SlideShare a Scribd company logo
Cytotoxic and Inflammatory Responses Induced by Outer Membrane
Vesicle-Associated Biologically Active Proteases from Vibrio cholerae
Ayan Mondal,a
Rima Tapader,a
Nabendu Sekhar Chatterjee,b
Amit Ghosh,a
* Ritam Sinha,c
Hemanta Koley,c
Dhira Rani Saha,d
Manoj K. Chakrabarti,a
Sun Nyunt Wai,e
Amit Pala
Division of Pathophysiology,a
Division of Biochemistry,b
Division of Bacteriology,c
and Histology and Electron Microscopy,d
National Institute of Cholera and Enteric
Diseases, Kolkata, India; Department of Molecular Biology, Umea Centre of Microbial Research, Umea University, Umea, Swedene
Proteases in Vibrio cholerae have been shown to play a role in its pathogenesis. V. cholerae secretes Zn-dependent hemaggluti-
nin protease (HAP) and calcium-dependent trypsin-like serine protease (VesC) by using the type II secretion system (TIISS). Our
present studies demonstrated that these proteases are also secreted in association with outer membrane vesicles (OMVs) and
transported to human intestinal epithelial cells in an active form. OMV-associated HAP induces dose-dependent apoptosis in
Int407 cells and an enterotoxic response in the mouse ileal loop (MIL) assay, whereas OMV-associated VesC showed a hemor-
rhagic fluid response in the MIL assay, necrosis in Int407 cells, and an increased interleukin-8 (IL-8) response in T84 cells, which
were significantly reduced in OMVs from VesC mutant strain. Our results also showed that serine protease VesC plays a role in
intestinal colonization of V. cholerae strains in adult mice. In conclusion, our study shows that V. cholerae OMVs secrete biolog-
ically active proteases which may play a role in cytotoxic and inflammatory responses.
Vibrio cholerae is the causative agent of the life-threatening dis-
ease cholera. Cholera epidemics in Haiti in 2010 provide evi-
dence that it still remains an ongoing public health threat (1).
Strains of the El Tor biotype O1 serogroup are responsible for
seventh pandemic and recent cholera outbreaks (2). Cholera toxin
(CT) and toxin-coregulated pilus (TCP) have been identified as
the major virulence factors for V. cholerae pathogenesis. CT is
responsible for profuse watery diarrhea, whereas TCP is essential
for sustaining colonization of human small intestine. V. cholerae
also secretes several proteases which may also play a role in its
pathogenesis (3).
The major protease secreted by V. cholerae strains is the 35-kDa
hemagglutinin protease (HAP) (4). HAP has been reported to
accelerate the bacterial detachment from cultured cells by diges-
tion of V. cholerae adhesins (5). HAP modulates the enterotoxige-
nicity of cholera toxin by nicking the A subunit of CT (6). HAP
also plays a role in processing hemolysin to its mature form by
removal of a 15-kDa N-terminal peptide (7). As shown in the
above-mentioned studies, HAP plays an indirect role in V. chol-
erae pathogenesis. The possibility of its direct role in pathogenesis
has been shown by Ghosh et al. (8). They reported that purified
HAP from a V. cholerae non-O1, non-O139 strain showed a hem-
orrhagic response in rabbit ileal loops (RILs) and an increase in
intestinal short-circuit current in Ussing’s chamber (8). Besides
HAP, the other major well-characterized metalloprotease is a 97-
kDa Vibrio cholerae protease, PrtV which has been shown to play a
role in virulence in the Caenorhabditis elegans infection model (9).
In an earlier study, we also reported the presence of a novel serine
protease encoded by VC1649 (VesC) in a hapA prtV mutant Vibrio
cholerae O1 strain and showed its role in the hemorrhagic re-
sponse in the rabbit ileal loop model (10).
Gram-negative bacteria, including V. cholerae, use 6 different
secretion systems (type I to type VI) to transport important viru-
lence factors to the cell envelope and the extracellular milieu (11).
Outer membrane vesicles (OMVs) may also serve as a mechanism
of delivering active virulence factors into host cells. The virulence
factors are protected from digestion by host proteases and are
secreted not individually but as multiple factors delivered in a
discrete package (12). Toxin delivery mediated by OMVs is recog-
nized as a potent virulence mechanism for many bacterial patho-
gens. Association of toxins with OMVs have been reported for
many pathogenic Gram-negative bacteria. Toxins such as ClyA
cytotoxin, ␣-hemolysin, and CNF1 of Escherichia coli, cytolethal
distending toxin (CDT) of Campylobacter jejuni, and vacuolating
cytotoxin A (VacA) of Helicobacter pylori are secreted through
OMVs (13, 14, 15, 16, 17). Bomberger et al. showed that multiple
virulence factors, such as ␤-lactamase, alkaline phosphatase, he-
molytic phospholipase C, and Cif, are directly delivered into the
host cytoplasm via fusion of OMVs with lipid rafts in the host
plasma membrane and that these toxins are rapidly distributed to
specific subcellular locations to affect host cell biology (18).
OMV-mediated secretion of biologically active CT, Vibrio chol-
erae cytolysin (VCC), and PrtV has been reported in V. cholerae
(19, 20, 21).
Recent studies on proteomic analysis of V. cholerae OMVs have
the shown presence of both HAP and the 59-kDa serine protease
VesC (22). However, the functions of these vesicle-associated pro-
teases have not been elucidated. Our study shows that V. cholerae
OMVs secrete biologically active proteases which may play a role
in cytotoxic and inflammatory responses.
Received 5 November 2015 Returned for modification 23 December 2015
Accepted 20 February 2016
Accepted manuscript posted online 29 February 2016
Citation Mondal A, Tapader R, Chatterjee NS, Ghosh A, Sinha R, Koley H, Saha DR,
Chakrabarti MK, Wai SN, Pal A. 2016. Cytotoxic and inflammatory responses
induced by outer membrane vesicle-associated biologically active proteases from
Vibrio cholerae. Infect Immun 84:1478–1490. doi:10.1128/IAI.01365-15.
Editor: A. Camilli
Address correspondence to Amit Pal, apal7364@rediffmail.com.
* Present address: Amit Ghosh, Department of Physiology, All India Institute of
Medical Sciences, Bhubaneswar, India.
Copyright © 2016, American Society for Microbiology. All Rights Reserved.
crossmark
1478 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
MATERIALS AND METHODS
Ethics statement. Animal experiments were performed after obtaining
necessary permission from the Institutional Animal Ethical Committee
(IAEC). The IAEC/CPCSEA approval number is APRO 7624/11/2010.
Bacterial strains. The bacterial strains, and oligonucleotides used in
this study are listed in Table 1. V. cholerae cells were routinely grown in
tryptic soy broth (TSB) (Difco, USA) at 37°C with shaking as described
earlier (10). E. coli TOP 10 cells were grown overnight in Luria broth (LB)
at 37°C with shaking at 200 rpm. Antibiotics were used at the following
concentrations: streptomycin, 100 ␮g/ml; kanamycin, 40 ␮g/ml; and am-
picillin, 100 ␮g/ml. Bacterial cells, including mutants, were maintained at
Ϫ70°C in LB containing 20% sterile glycerol.
Complementation of the VC1649 gene in V. cholerae strain CHA6.8
⌬prtV ⌬VC1649. The VC1649 gene was amplified from genomic DNA
isolated from CHA6.8 ⌬prtV using VesC forward and reverse primers
(Table 1). Cloning was performed as described by Gosink et al. (23). The
amplified VC1649 gene was cloned into the pBAD-TOPO TA (Invitrogen,
USA) cloning vector under the control of an arabinose-inducible pro-
moter using a cloning kit from Invitrogen and was transformed in E. coli
TOP 10 cells. Transformed colonies (Ampϩ
) were selected in a Luria agar
(LA) plate containing 100 ␮g/ml ampicillin. Positive clones were selected
from the transformed colonies by colony PCR with the pBAD forward and
VesC reverse primers supplied in the cloning kit, and plasmids were iso-
lated using a plasmid isolation kit (Thermo Fisher, USA). Plasmid
pVC1649 was transformed into V. cholerae strain CHA6.8 ⌬prtV
⌬VC1649 by electroporation (23).
IsolationandpurificationofOMVsfromV.choleraestrains.V.chol-
erae cells were grown in 2 liter of TSB for 18 h at 37°C under shaking
conditions, and outer membrane vesicles (OMVs) were prepared from
late-log-phase culture as described previously (17). Crude vesicles were
isolated from strain Comp VC1649 grown in 250 ml of TSB using L-ara-
binose induction as described by Chen et al. (24). Isolated crude OMVs
from C6709, OMVs from its isogenic protease mutants, and OMVs from
Comp VC1649 were purified by density gradient centrifugation on a dis-
continuous Optiprep-iodixanol (Sigma, USA) gradient as described by
Elluri et al. (20). Fractions of equal volumes were sequentially removed
after density gradient centrifugation and analyzed by Western blotting
using anti-OmpU antibody. OmpU is the marker protein for V. cholerae
OMVs. The protein content of purified OMVs was estimated using the
Bradford assay.
Transmission electron microscopy (TEM). Twenty micrograms of
purified OMVs was placed on carbon-coated nickel grids (300 mesh)
(Sigma, USA), stained with 2% aqueous uranyl acetate, rinsed, and exam-
ined with a transmission electron microscope operating at 60 kV (FEI
Tecnai 12; Bio, Twin, The Netherlands).
RaisingofantiseraagainstHAP,VesC,andOMVs.HAPwaspurified
from the culture supernatant of C6709 as described by Ghosh et al. (8).
Antiserum to purified HAP was prepared by immunizing a New Zealand
White rabbit by intramuscular injection with 100 ␮g of HAP emulsified
with an equal volume of Freund’s complete adjuvant (Sigma, USA). This
was followed by four booster injections with 100 ␮g of HAP and incom-
plete adjuvant (Sigma, USA) at 7-day intervals. Blood samples were col-
lected from rabbits on day 0 and 3 days after the final injections and were
allowed to clot at room temperature for 30 min. Sera were collected,
followed by centrifugation (1,000 rpm, 10 min), dilution in phosphate-
buffered saline (PBS) (Sigma, USA), and storage at Ϫ80°C (25). Antise-
rum against the 59-kDa serine protease (VesC) was also raised in a New
Zealand White rabbit. VesC was partially purified as described earlier
(10). The mass spectrometrically identified band of VesC in a native poly-
acrylamide gel was excised, homogenized in PBS, and administered into
an intramuscular region of a New Zealand White rabbit. Antiserum
against VesC was drawn from the rabbit after four booster injections as
described above. Antiserum against OMVs was prepared by immunizing
7-week-old mice (BALB/c). Mice were immunized subcutaneously with
50 ␮g of OMVs isolated from C6709 at the first, second, and third weeks,
followed by a booster dose of 75 ␮g of the OMVs at the fourth week. The
serum was collected, and the antibody titer was determined by enzyme-
linked immunosorbent assay (ELISA) with the preimmune serum as a
control.
SDS-PAGE and Western blot analysis. V. cholerae OMVs were sepa-
rated by 10% SDS-PAGE. Briefly, 20 ␮g each of purified OMVs and
whole-cell lysates of C6709 and CHA6.8 were mixed with Laemmli sample
TABLE 1 Bacterial strains, plasmids, and primers
Strain, plasmid, or primer Relevant characteristics
Source or
reference
Vibrio cholerae strains
C6709 Wild type (O1 El Tor); Smr
56
CHA6.8 C6709 ⌬hapA::Kan; Smr
Kmr
10
CHA6.8 ⌬prtV CHA6.8 ⌬prtV; Smr
Kmr
10
CHA6.8 ⌬prtV ⌬VC1649 (VesC mutant) CHA6.8 ⌬prtV ⌬VC1649; Smr
Kmr
10
Comp VC1649 CHA6.8 ⌬prtV ⌬VC1649::pVC1649; Smr
Kmr
Ampr
This study
E. coli strain TOP 10 FϪ
mcrA ⌬(mrr-hsdRMS-mcrBC) ␾80lacZ⌬M15⌬lacX74 recA1
araD139 ⌬(ara-leu)7697 galU galK rpsL (Strr
) endA1 nupG
Invitrogen, USA
Plasmids
pBAD TOPO-TA Ampϩ
TA cloning vector plasmid Invitrogen, USA
pVC1649 pBAD TOPO with Vibrio cholerae VC1649 (VesC) This study
Primers
IL 8 F1 5=-ATGGGGAAGGTGAAGGTCGG-3= 57
IL 8 R1 5=-TCTCAGCCCTCTTCAAAAACTTCTC-3= 57
GAPDH F1 5=-ATGGGGAAGGTGAAGGTCGG-3= 58
GAPDH R1 5=-GGATGCTAAGCAGTTGGT-3= 58
pBAD F 5=-ATGCCATAGCATTTTTATCC-3= Invitrogen, USA
pBAD R 5=-GATTTAATCTGTATCAGG-3= Invitrogen, USA
VesC F 5=-ATGAACAAAACATTTTTATC-3= This study
VesC R 5=-TCAGACGCGTTGACGACG-3= This study
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1479Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
buffer (1:1) and boiled for 10 min prior to electrophoresis. Proteins with
known molecular weights (Thermo Fisher, USA) were used as markers.
Gels were stained with Coomassie brilliant blue stain after electrophoresis.
For immunoblotting, the separated proteins were transferred onto a ni-
trocellulose membrane (Bio-Rad, USA) by the wet transfer method using
a Trans-Blot apparatus (Bio-Rad, USA). The membrane was blocked at
room temperature for 1 h in 5% nonfat dry milk powder (Bio-Rad, USA)–
PBS–Tween 20. After incubation, the membrane was washed and incu-
bated with rabbit anti-HAP (1:1,000 dilution in 5% [wt/vol] nonfat dry
milk), and anti-VesC (1:10,000 dilution in 5% [wt/vol] nonfat dry milk)
antisera, followed by incubation for 1 h at room temperature with a
1:10,000 dilution of anti-rabbit horseradish peroxidase-conjugated sec-
ondary antibody (Thermo Fisher, USA). The blots were visualized with
ECL Western blotting detection reagent (Millipore, USA).
Proteolytic digestion of prohemolysin. Proteolytic digestion of El
Tor prohemolysin was performed as described earlier (7). Briefly, 10 ␮l of
79-kDa prohemolysin (1 mg/ml) was mixed with 10 ␮l (100 ␮g/ml) of
purified C6709 OMVs and incubated at 37°C for 30 min, and SDS-PAGE
was performed. Untreated purified prohemolysin and HAP-treated pro-
hemolysin were used as negative and positive controls, respectively.
Proteinase K susceptibility assay. The proteinase K susceptibility as-
say was performed as described previously (26). Briefly, OMVs were
treated with proteinase K (1 ␮g/ml) for 30 min at 37°C in either the
presence or absence of 1% SDS, and samples were analyzed by immuno-
blotting against anti-HAP and anti-VesC antibodies raised in rabbit.
Media for cell lines and culture conditions. Int407 and T84 human
intestinal epithelial cells were used in this study. Int407 cells were grown in
25-ml tissue culture flasks (BD Biosciences) containing minimal essential
medium (MEM) (Gibco, USA) supplemented with 10% fetal bovine se-
rum (Gibco, USA) along with penicillin G and streptomycin sulfate
(Sigma, USA) at 37°C in a CO2 incubator (Heraeus, Germany). Cells at 80
to 90% confluence were seeded in 6-well and 24-well tissue culture plates
(BD Biosciences) at a concentration of approximately 105
cells/well. Cells
were permitted to reach confluence by allowing them to grow for 24 h in
a CO2 incubator. T84 cells were used for interleukin-8 (IL-8) assay, and
the cells were maintained in Dulbecco’s modified Eagle’s Medium-nutri-
ent F12 (DMEM-F12) (Gibco, USA) supplemented with antibiotic solu-
tion as described above.
Tissue culture assay. The tissue culture assay was performed as de-
scribed by Ghosh et al. (8). Briefly, OMVs from C6709 and its isogenic
knockout strains were filter sterilized and serially diluted in serum-free
MEM from 0.5 ␮g/ml to 100 ␮g/ml. The exhaust media were aspirated
from 24-well tissue culture plates containing Int407 cells grown to con-
fluence and washed with PBS, and serially diluted OMVs were added and
grown at 37°C in a CO2 incubator for 24 h. Purified HAP was used as
positive control. Results were analyzed by phase-contrast microscopy and
photographs taken (CK 40; Olympus).
OMV internalization assay. Int407 cells were incubated with 20
␮g/ml of purified OMVs from C6709 and its isogenic protease mutants
for 1 h at 37°C in a 5% CO2 atmospheric chamber to monitor internaliza-
tion of V. cholerae OMVs. The effects of several endocytic inhibitors (nys-
tatin, dynasore, and methyl-␤-cyclodextrin [M␤CD]) on OMV uptake
were examined by pretreating Int407 cells with nystatin (25 ␮g/ml) and
dynasore (80 ␮M) for 1 h and with M␤CD (1 mg/ml) (Sigma, USA) for 30
min as described by Parker et al. (27). This was followed by incubation
with purified OMVs for 1 h. Cells were then fixed with 4% paraformalde-
hyde at 4°C for 20 min, followed by permeabilization with 1% Triton
X-100 in PBS for 20 min at room temperature. Mouse antiserum against
OMVs (1:2,000) was added to OMV-treated cells and incubated overnight
at 4°C in a moist chamber. Cells were washed with sterile PBS and incu-
bated with tetramethyl rhodamine isocyanate (TRITC)-conjugated anti-
mouse secondary antibody (1:500) and were kept in the dark at 37°C for
90 min.
For colocalization studies, primary antibodies against HAP (1:1,000)
and OMVs (1:2,000) raised in rabbit and mouse, respectively, were added
to C6709 and CHA6.8 OMV-treated cells. Primary antibodies against
VesC (1:2,500) and OMVs (1:2,000) were added to cells treated with
OMVs from C6709, isogenic protease mutants of C6709, and the VC1649
complemented strain and incubated overnight as described above. Cells
were incubated with fluorescein isothiocyanate (FITC)-conjugated anti-
rabbit IgG (1:500) and TRITC-conjugated anti-mouse antibody (1:500)
and were kept in the dark at 37°C for 90 min after incubation. Cells were
washed and resuspended in 100 ␮l of PBS. Approximately 20 ␮l of resus-
pended sample was used to make a thin film on a glass slide. One drop of
90% glycerol was added, a thin glass coverslip was placed on it, and the
slide was viewed under a confocal microscope (model LSM 510 META;
Zeiss, Germany) and excited at 488 nm and 543 nm for FITC and TRITC,
respectively. The emission filters used were 475 nm to 525 nm for FITC
and 558 nm to 601 nm for TRITC. Experiments were also done with PBS
as negative control. Merged images were developed using Image J soft-
ware.
Mouse intestinal fluid accumulation assay. The mouse intestinal
fluid accumulation assay was performed as described earlier (28). Briefly,
20 ␮g of purified OMVs from V. cholerae and its isogenic protease mutant
strains was injected into ligated intestinal segments of about 4 cm in
length from 4- to 5-week-old adult BALB/c mice. All experimental proce-
dures were performed under anesthesia with ketamine. After 6 h, mice
were sacrificed and loops were excised. The enterotoxic activity was de-
termined by measuring the fluid accumulation (FA) ratio, which is the
ratio of fluid accumulated in the intestinal loop to the length of the loop
(g/cm). PBS was used as a negative control. Values were expressed as
mean Ϯ standard deviation (SD) of the mean (n ϭ 5 mice per group).
Histopathological studies of mouse ileal loops (MILs). Mouse ileal
tissues were collected and fixed in 10% neutral buffered formalin solution
for histopathological studies. The tissues were further embedded in par-
affin and processed following the standard protocol. Three- to four-mi-
crometer thin sections were cut in a microtomy rotor (Leica, Germany).
The sections were strained with hematoxylin and eosin and observed un-
der a light microscope. Photographs were taken under a magnification of
ϫ40 with a Leica DMLB microscope (Solms, Germany) equipped with a
digital imaging system.
Flow cytometric analysis. Flow cytometric analysis was performed as
described by Thay et al. (29). Briefly, Int 407 cells were treated with OMVs
from V. cholerae strain C6709 and its protease knockout mutant strains at
concentrations of 20 ␮g/ml and 40 ␮g/ml for 4 h and then stained with
fluorescein isothiocyanate (FITC)-conjugated annexin V and propidium
iodide (PI) (BD Pharmingen) according to the manufacturer’s instruc-
tions. The cells were stained with FITC-annexin V in annexin V binding
buffer for 10 min, followed by addition of PI, and analyzed by flow cy-
tometry using Cell Quest Pro software (BD Biosciences). A total of 104
cells were acquired for data analysis in each set of experiments. Values
were expressed as mean Ϯ SD of the mean for 3 different observations
(n ϭ 3).
CT-bead ELISA. CT concentrations in OMVs and culture superna-
tants of C6709 and its isogenic protease mutants grown in TSB were mea-
sured by bead ELISA as described by Oku et al. (30). Culture supernatant
of V. cholerae N16961 grown in Syncase medium was used as a positive
control.
Mouse intestinal colonization assay. Pathogen-free adult 4- to
5-week-old BALB/c mice were used for the colonization assay. Prior to the
experiment, all mice were fasted for 24 h with free access to sterile water.
The experiment was performed as described by Olivier et al. (31). Briefly,
mice were injected intraperitoneally with a mixture of 35 mg/kg ketamine
and 5 mg/kg xylazine for anesthesia. Two hundred microliters of suspen-
sions of ϳ109
viable bacterial cells was administered orally with two bo-
luses of 0.5 ml of 5% (wt/vol) NaHCO3. All mice were kept in microiso-
lator cages with free access to food and sterile water after inoculation. Mice
were euthanized with Euthanasia 6 solution after 18 h of oral inoculation.
The small intestine above the cecum was collected after dissection, ho-
mogenized, and then serially diluted in sterile PBS (pH 7.4). Homoge-
Mondal et al.
1480 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
nized materials were plated on appropriate antibiotic-containing tryptic
soy agar (TSA) plates and incubated for 24 h. All experiments were per-
formed in triplicates, and values were expressed as mean Ϯ SD (n ϭ 10
mice per group) (*, P Ͻ 0.05; **, P Ͻ 0.01) and compared with the CFU
obtained for C6709.
ELISA and semiquantitative RT-PCR for IL-8 estimation. For en-
zyme-linked immunosorbent assays (ELISAs), the growth medium was
removed from the plates. Cells were washed with PBS (Sigma, USA) and
maintained in antibiotic- and serum-free DMEM-F12. The cells were
treated with 10 ␮g/ml of V. cholerae OMVs from C6709 and its isogenic
knockout mutant strains for IL-8 stimulation. Supernatants were col-
lected after 24 h and centrifuged to remove dead cells, and IL-8 concen-
trations were determined by ELISA using an ELISA kit from Invitrogen
(USA) according to the instructions of the manufacturer. For reverse
transcription-PCR (RT-PCR), total RNA was isolated from the OMV-
treated T84 cells using TRIzol reagent (Invitrogen, USA). RNA was quan-
tified by spectrophotometric analysis and the quality of the RNA samples
determined by estimating the A260/A280 ratio. cDNA was prepared from
1.5 ␮g of total RNA using the Superscript II first-strand synthesis system
for RT-PCR (Invitrogen, USA). A 2.5-␮l portion of cDNA was PCR am-
plified in a 25-␮l reaction volume containing 10 mM Tris-Cl (pH 8.3), 50
mM KCl, 2.0 mM MgCl2, 2.5 mM each deoxynucleoside triphosphate
(dNTP), 20 pmol of each primer, and 1 U of Taq DNA polymerase
(Roche, Germany). The primers used for IL-8 and GAPDH (glyceralde-
hyde-3-phosphate dehydrogenase) are listed in Table 1. The PCR condi-
tions were as follows: an initial denaturation step at 95°C for 5 min fol-
lowed by 35 cycles of 95°C (1 min), 55°C and 60°C for IL-8 and GAPDH,
respectively (1 min), and 72°C (30 s). The final extension step was done at
72°C for 7 min. The products were electrophoresed on a 2% agarose gel,
stained with ethidium bromide, and visualized using a gel documentation
system (Bio-Rad, USA). The data from ELISA and RT-PCR was recorded
as mean Ϯ SD from at least three groups of experiments. Densitometric
quantification of RT-PCR bands was performed with Quantity One soft-
ware (Bio-Rad, USA).
Statistical analysis. All experiments were performed in triplicates.
Values were expressed as mean Ϯ standard deviation of the mean. Vari-
ables were compared for significance using two-way analysis of variance
and the Bonferroni test, with one asterisk indicating a P value between
0.01 to 0.05 and two asterisks indicating a P value between 0.01 to 0.001.
RESULTS
Fractions of HAP and VesC are associated with OMVs of Vibrio
cholerae C6709 and hapA mutant strains, respectively. V. chol-
erae secretes abundant vesicles during its normal course of growth
(32). To investigate the association of proteases with outer mem-
brane vesicles (OMVs), crude OMVs were isolated from culture
supernatants of the pathogenic O1 El Tor strain C6709 and its
protease mutants by ultracentrifugation, followed by purification
using Optiprep density gradient centrifugation. Fractions ob-
tained from the gradient centrifugation of OMVs were analyzed
by immunoblotting using OmpU antiserum. Fractions showing
reactivity with OmpU antiserum were examined by transmission
electron microscopy for ultrastructural analysis and used for fur-
ther experiments. OMVs of C6709 appeared as spherical electron-
dense materials of heterogeneous sizes with a diameter in the
range of 25 to 150 nm. Major morphological differences were not
observed in OMVs from the isogenic protease mutant strains and
OMVs from Comp VC1649. To investigate OMV-associated se-
cretion of HAP, OMVs from HAP-producing C6709 and ⌬hapA
strain CHA6.8 were subjected to SDS-PAGE along with whole-cell
lysates from the respective strains, and immunoblotting was per-
formed using polyclonal antiserum against HAP. Concentrated
OMV-free culture supernatants from C6709 and purified HAP
were used as positive controls. An immunoreactive band corre-
sponding to HAP was observed only in C6709 OMVs. The pres-
ence of HAP was not observed in whole-cell lysate prepared from
strain C6709. The immunoblot results showed the presence of the
mature 45-kDa form of HAP in OMVs instead of its processed
35-kDa form, which was present in OMV-free culture supernatant
of C6709 (Fig. 1B). The presence of VesC in the OMVs was de-
tected by immunoblotting using VesC antiserum (1:10,000). VesC
was detected in the OMVs from the CHA6.8 and CHA6.8 ⌬prtV
strains and the VC1649 complemented construct. In contrast, its
presence was not observed in OMVs from C6709 and CHA6.8
⌬prtV ⌬VC1649 (Fig. 1C). The above results demonstrated that
FIG 1 Association of HAP and VesC with V. cholerae OMVs by immunoblot analysis. (A) Ten percent SDS-PAGE profile of the OMVs of Vibrio cholerae strains
C6709 (lane 3), CHA6.8 (lane 4), CHA6.8 ⌬prtV (lane 5), CHA6.8 ⌬prtV ⌬VC1649 (lane 6), and Comp VC1649 (complemented) (lane 7). Five micrograms of
purified HAP was resolved in lane 2, and a prestained protein ladder (Thermo Fisher, USA) is in lane 1. (B) Immunoblot analyses with OMVs from C6709 and
CHA6.8 and whole-cell lysates from the respective strains against HAP antiserum. Purified 45-kDa and 35-kDa HAP and concentrated OMV-free culture
supernatant of C6709 were used as positive controls. (C) Immunoblot analyses with OMVs of C6709 and its isogenic protease mutants against VesC antiserum.
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1481Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
HAP is secreted in association with OMVs from the V. cholerae
wild-type strain C6709, whereas the serine protease VesC is se-
creted in association with OMVs from ⌬hapA strains.
LocalizationofproteasewithintheOMVs.Localizationofthe
HAP and VesC within the OMVs was confirmed by proteinase K
susceptibility assay. Clear proteolytic digestion of HAP was ob-
served only when OMVs from C6709 were incubated with protei-
nase K in the presence of 1% SDS, and this proteolytic digestion
was inhibited in the presence of phenylmethylsulfonyl fluoride
(PMSF) (Fig. 2A). A similar result was observed with OMV-asso-
ciated VesC in the CHA6.8 ⌬prtV strain (Fig. 2B). These results
confirmed the luminal localization of HAP and VesC within the
vesicles.
Protease-associated OMVs bind to and are internalized by
human intestinal epithelial cells in a protease-independent
manner. The interaction of OMVs with Int407 cells was studied
by confocal microscopy. OMV-specific red fluorescence was ob-
served throughout Int407 cells treated with OMVs from C6709
and its isogenic protease mutant strains (Fig. 3A, panels 4 and 7,
and C, panels 1, 4, 7, and 10). In contrast, no fluorescence was
observed in cells treated with buffer (Fig. 3A, panel 1). Internal-
ization of OMVs in Int407 cells was confirmed by treatment with
different endocytic inhibitors prior to addition of OMVs. The
cholesterol-sequestering agent M␤CD inhibited the entry of V.
cholerae OMVs in Int407 cells (Fig. 3B, panels 5 and 6). This result
suggests that V. cholerae OMVs may be internalized into the cells
by cholesterol-enriched lipid rafts. M␤CD has also been reported
to inhibit clathrin-mediated endocytosis when applied at high
concentrations (33). Inhibition of vesicular endocytosis was fur-
ther confirmed by using nystatin, a cholesterol binding agent,
which disrupts lipid rafts but does not affect clathrin-mediated
endocytosis. Our results showed that nystatin also inhibits the
entry of V. cholerae OMVs in Int407 cells (Fig. 3B, panels 7 and 8).
Interestingly, dynasore (an inhibitor of the dynamin-dependent
endocytic pathway) failed to inhibit internalization of OMVs into
Int407 cells (Fig. 3B, panels 9 and 10). The inhibition of internal-
ization of C6709 OMVs is shown in Fig. 3B, which shows that
C6709 OMVs failed to enter Int407 cells upon treatment with
M␤CD and nystatin. The above results suggested that V. cholerae
OMVs are internalized into In407 cells by lipid raft-dependent
endocytosis.
To investigate OMV-mediated transport of HAP in Int407
cells, confocal microscopy was performed, using HAP antiserum
and FITC-conjugated secondary antibody. The merged images
showed colocalization of HAP and OMVs in Int407 cells when
treated with HAP-associated C6709 OMVs (Fig. 3A, panel 6),
whereas cells treated with CHA6.8 showed only OMV-specific red
fluorescence (Fig. 3A, panel 9). OMVs were labeled with VesC
antiserum and FITC-conjugated secondary antibody to assess
whether VesC is transported into cells. Our results with the colo-
calization assay clearly showed that VesC is transported into
Int407 cells through OMVs from hapA mutant strains CHA6.8
and CHA6.8 ⌬prtV (Fig. 3C, panels 6 and 9). Colocalization of
OMVs and VesC was not observed when cells were treated with
CHA6.8 ⌬prtV ⌬VC1649 OMVs. However, the merged image
showed adherence and internalization of OMVs in the absence of
VesC (Fig. 3C, panel 12), and this suggests that VesC is not respon-
sible for adherence of OMVs in Int407 cells. OMV-mediated
transport of VesC was further confirmed with OMVs from strain
Comp VC1649, which showed both OMV- and VesC-specific sig-
nals in the cells (Fig. 3C, panels 13 and 14).
OMV-associated proteases are biologically active. Studies
have shown that OMVs deliver cargo to the specific targets in an
active form (34). In the present study, we showed that both HAP
and VesC are transported into intestinal epithelial cells through
OMVs of their producing strains. HAP-associated C6709 OMVs
induced morphological changes of Int407 cells in a dose-depen-
dent manner when treated for 24 h (Fig. 4A, panel I). C6709
OMVs showed a cell-distending effect at a low concentration (20
␮g/ml), whereas they showed a cell-rounding effect similar to that
for purified HAP at a higher concentration (50 ␮g/ml) (Fig. 4A,
panel I). In contrast, such morphological changes were not ob-
served when cells were treated with OMVs from CHA6.8 and
other protease mutant strains. The proteolytic activity of HAP-
associated OMVs was studied on 79-kDa prohemolysin to con-
firm its biological activity. HAP has been reported to proteolyti-
cally cleave prohemolysin by digestion of the N-terminal
polypeptide chain to its 65-kDa mature form (7). Our results
showed that OMV-associated HAP digests 79-kDa prohemolysin
to produce 65-kDa and 50-kDa fragments, similar to the effect
observed with purified HAP (Fig. 4A, panel II). Our results con-
firmed that OMV-associated HAP is biologically active. In earlier
studies, we have reported that purified HAP and partially purified
VesC induce a hemorrhagic fluid response in the RIL assay (8, 10).
The biological activities of OMV-associated proteases were evalu-
ated in vivo by fluid accumulation assay in the ilea of adult mice.
OMVs from wild-type strain C6709 showed significant fluid ac-
cumulation (FA ratio, 0.20 Ϯ 0.03; n ϭ 5) (**, P Յ 0.01) compared
to the control (PBS treated) (Fig. 4B, panel I). Histopathological
studies of ileal tissues revealed widely dilated and ruptured villi
and the presence of inflammatory cells in the mucosa (magnifica-
tion, ϫ40) (Fig. 4B, panel IIB). Interestingly, hemorrhagic fluid
accumulation was observed in the intestinal segments when they
were treated with OMVs from CHA6.8 and CHA6.8 ⌬prtV. His-
topathological studies of the ileal tissues showed an altered villous
structure and mild hemorrhage in the submucosal membrane
when treated with CHA6.8 OMVs (Fig. 4B, panel IIC). However,
CHA6.8 ⌬prtV OMVs caused maximum damage of the ileal tis-
sues (Fig. 4B, panel IID). The ileal tissues showed grossly dis-
FIG 2 Localization of proteases in OMVs by proteinase K susceptibility assay.
OMVs from C6709 and CHA6.8 ⌬prtV were incubated at 37°C for 30 min with
or without 1 ␮g/ml proteinase K alone or with 1% SDS or 1 mM PMSF. Lane
1, control reaction; lane 2, sensitivity to extracellular protease; lane 3, inhibi-
tion of extracellular proteases; lane 4, protease sensitivity to luminal proteins;
lane 5, inhibition of protease activity by PMSF (control). OMVs were incu-
bated under the above-described conditions, precipitated with acetone, dried,
electrophoresed, and immunoblotted against HAP (A) or VesC (B) antiserum.
Mondal et al.
1482 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
FIG 3 Colocalization of proteases and OMVs in the Int407 cell line. (A) Int407 cells were treated with OMVs from strain C6709 (panels 4, 5, and 6) or CHA6.8
(panels 7, 8, and 9) or with sterile PBS as a negative control (panels 1, 2, and 3), incubated with HAP and OMV antisera, and stained with TRITC (red)-conjugated
anti-mouse antibody (panels 1, 4, and 7). Red fluorescence was detected in all OMVs except the negative control. HAP was detected using anti-HAP and
FITC-conjugated (green) anti-rabbit antibody in panel 5. Merged images are shown in panels 3, 6, and 9. The nucleus was stained with DAPI (4=,6=-diamidino-
2-phenylindole). (B) Inhibition of internalization of C6709 OMVs in Int407 cells treated with dynasore, methyl-␤-cyclodextrin (M␤CD), and nystatin prior to
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1483Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
rupted villi with hemorrhage in all layers of the mucosa. In con-
trast, ileal tissues treated with CHA6.8 ⌬prtV ⌬VC1649 OMVs
showed an almost normal villous structure with minimum hem-
orrhage in the mucosa and submucosa in histopathological stud-
ies (Fig. 4B, panel IIE). Fluid accumulation in the intestinal seg-
ments was also significantly reduced (FA ratio, 0.07 Ϯ 0.03; n ϭ
5). These results may suggest that OMV-associated VesC may be
responsible for the hemorrhagic fluid response in the mouse ileal
loop assay. The MIL assay with OMVs from Comp VC1649
showed restoration of hemorrhagic fluid in intestinal segments,
and histopathological studies showed mucosal damage and the
presence of inflammatory cells in the mucosa and submucosa (Fig.
4B, panel IIF). The presence of VesC in the OMVs may responsible
for the hemorrhagic fluid responses. However, it was reported
earlier that CT is responsible for fluid accumulation in the mouse
ileal loop assay (35). We measured the CT titers of V. cholerae
OMVs and culture supernatants by CT-bead ELISA to confirm
whether the OMV-mediated inflammatory responses is due to the
presence of CT in the OMVs. Both cell-free culture supernatants
and OMVs from V. cholerae strains grown in TSB showed almost
negligible CT titers compared to that of the positive-control strain
N16961 grown in Syncase medium (Fig. 4C) Therefore, the hem-
orrhagic response by OMVs in the MIL assay is not due to CT. The
above results confirmed that OMV-associated VesC is responsible
for hemorrhagic fluid accumulation in mouse ileal loops.
Host cell death induced by V. cholerae OMVs is dependent
on proteases. The cell death mechanisms induced by V. cholerae
OMVs were analyzed by flow cytometry. Flow cytometric analysis
with C6709 OMVs showed a dose-dependent apoptotic response
in Int407 cells (annexin Vϩ
/PIϪ
fraction). More than 50% cell
death was observed at 20 ␮g/ml of C6709 OMVs compared to the
untreated control. However, CHA6.8 OMVs induced greater cy-
totoxicity than C6709 OMVs, and cell death occurred by both
apoptosis and necrosis. Interestingly 30% reduced cell death was
observed when cell were treated with CHA6.8 ⌬prtV OMVs com-
pared to CHA6.8 OMVs. However, a 2-fold increase in the ne-
crotic cell population was observed compared to that with
CHA6.8 OMVs (Fig. 5C). CHA6.8 ⌬prtV ⌬VC1649 OMVs
showed reduced cytotoxicity compared to C6709, CHA6.8, and
CHA6.8 ⌬prtV OMVs. OMVs from the VC1649 complemented
strain restored the cytotoxicity in Int407 cells. These results clearly
demonstrated that VesC is responsible for necrotic damage of
Int407 cells (Fig. 5B and C).
VesC plays a role in intestinal colonization of V. cholerae in
the adult mouse model. Assessment of colonization of V. cholerae
strains in mouse intestinal epithelium showed significantly re-
duced bacterial colonization of the triple knockout mutant strain
CHA6.8 ⌬prtV ⌬VC1649 (CFU 3 ϫ 103
cells/g of intestine) com-
pared with the ⌬hapA ⌬prtV strain CHA6.8 ⌬prtV (CFU 8 ϫ 105
cells/g of intestine) (Fig. 6). The ⌬hapA strain CHA6.8 showed
increased colonization (CFU 1 ϫ 108
cells/g of intestine) com-
pared to wild-type strain C6709 (CFU 3 ϫ 107
cells/g of intestine).
There was a significant decrease in colonization in the CHA6.8
⌬prtV ⌬VC1649 strain compared to the CHA6.8 ⌬prtV strain.
Interestingly, when the VC1649 gene was complemented in
CHA6.8 ⌬prtV ⌬VC1649, the strain showed 3-fold-increased col-
onization. These results suggest that VesC may play a role in col-
onization in adult mice.
OMV-mediated IL-8 secretion from T84 cells is dependent
on VesC. OMVs isolated from C6709 and its protease mutant
Vibrio cholerae strains were incubated with T84 cells for 24 h, and
culture supernatants were taken and assayed for IL-8 expression
by ELISA and semiquantitative RT-PCR. Significant IL-8 mRNA
expression was observed when cells were treated with CHA6.8,
CHA6.8 ⌬prtV, and Comp VC1649, as visible IL-8-specific bands
were observed in agarose gel electrophoresis. OMVs from the
CHA6.8 ⌬prtV and Comp VC1649 strains induced maximum
mRNA expression (Fig. 7B). Protein expression in the same set of
experiments was studied by IL-8 ELISA to assess whether the IL-8
mRNA level is correlated at the protein level. The ELISA results
showed that OMVs from CHA6.8 induced more than a 2.5-fold
increase in IL-8 secretion compared to those from wild-type strain
C6709 and a ϳ10-fold increase compared to those from untreated
cells. Interestingly, IL-8 secretion was increased ϳ25-fold com-
pared to that in untreated cells when cells were treated with
CHA6.8 ⌬prtV OMVs in 24 h. This increased IL-8 secretion was
reduced 4-fold when cells were treated with OMVs from the VesC
mutant strain (Fig. 7A). Upon VesC complementation in the
hapA prtV VC1649 triple knockout mutant strain, IL-8 induction
by OMVs was significantly increased. These results suggest that
VesC may act as an inducer for OMV-mediated IL-8 secretion in
human intestinal epithelial cells.
DISCUSSION
Proteases produced by microorganisms play an important role in
virulence (3). Vibrio cholerae secretes hemagglutinin protease
(HAP), V. cholerae protease (PrtV), and serine proteases through
the type II secretion system (TIISS) (21, 36). Earlier we have re-
ported that purified HAP from a ctx-negative V. cholerae non-O1,
non-O139 strain and VesC from a ⌬hapA ⌬prtV Vibrio cholerae
strain may play a role in pathogenesis by inducing a hemorrhagic
response in the rabbit ileal loop (RIL) assay (8, 10). Our present
study demonstrated that HAP and VesC are also secreted in asso-
ciation with outer membrane vesicles (OMVs). Our results
showed that HAP is secreted in association with OMVs from wild-
type strain C6709, whereas the 59-kDa serine protease VesC is
secreted in association with OMVs from hapA mutant strains but
not from wild-type strain C6709. The absence of VesC in C6709
OMVs could be due to its proteolytic degradation in the presence
of HAP, as it has been reported to mask the secretion of other
proteases from V. cholerae (3, 10). Earlier studies have shown that
secretion of serine proteases is regulated by metalloproteases in
Vibrio species (3, 37). Syngkon et al. showed that 90% of the pro-
tease activity in strain C6709 is due to HAP, as only residual pro-
addition of OMVs. Cells with untreated OMVs were used as a negative control and cells without inhibition as a positive control. OMVs were labeled with
TRITC-conjugated antibody, and the nucleus was stained with DAPI. (C) Int407 cells were treated with C6709 OMVs (panels 1, 2, and 3), CHA6.8 OMVs (panels
4, 5, and 6), CHA6.8 ⌬prtV OMVs (panels 7, 8, and 9), CHA6.8 ⌬prtV⌬ VC1649 OMVs (panels 10, 11, and 12), and Comp VC1649 (panels 13, 14, and 15),
incubated with VesC and OMVs antisera, and stained with red TRITC-conjugated anti-mouse antibody (panels 1, 4, 7, 10, and 13). Red fluorescence were
detected in all OMVs. The presence of VesC was detected using anti-VesC and FITC-conjugated (green) anti-rabbit antibodies (panels 5, 8, and 14). Merged
images are shown in panels 3, 6, 9, 12, and 15. The nucleus was stained with DAPI.
Mondal et al.
1484 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
FIG 4 Protease-associated OMVs are biologically active. (A) Biological activities of HAP-associated OMVs. Panel I, Int407 cells were treated with OMVs from
C6709 at 50 ␮g/ml and 20 ␮g/ml and OMVs from CHA6.8 for 24 h at 37°C. The higher concentration of C6709 OMVs showed a cell-rounding effect, whereas
the lower concentration showed a cell-distending effect. No morphological changes were observed in CHA6.8. Panel II, immunoblot analyses against hemolysin
antiserum with 10 ␮g purified hemolysin treated with HAP and C6709 OMVs and purified hemolysin as positive control. (B) Panel I, effect of OMVs of Vibrio
cholerae strains in the mouse ileal loop assay. Twenty micrograms (100 ␮g/ml) of OMVs from wild-type strain C6709 and its knockout derivatives was introduced
into ligated ileal loops of adult mice. The mice were sacrificed after 6 h, loops were excised, and fluid accumulation was calculated as the loop weight/length ratio.
Images of mouse ileal loops treated with V. cholerae OMVs are shown. The graph shows a summary of the data, represented as mean Ϯ SD (n ϭ 5 mice per group).
Variables were compared for significance using two-way analysis of variance and the Bonferroni test (*, P value between 0.01 and 0.05; **, P value between 0.01
and 0.001). Panel II, histopathological studies of mouse ileal loops. Twenty micrograms of OMV-treated ileal tissues was processed for histopathological analysis,
and photomicrographs were taken at a magnification of ϫ40. A, PBS-treated ileal tissues showed normal villi with mucosal structure; B, a magnified image of ileal
tissues treated with C6709 OMVs shows dilated and ruptured villi with accumulation of inflammatory cells in mucosal layers; C, CHA6.8 shows an altered villous
structure with mild hemorrhage in the submucosa; D, CHA 6.8 ⌬prtV OMV-treated ileal tissues show grossly disrupted villi with hemorrhage in all layers of the
mucosa; E, ileal tissues treated with OMVs of strain CHA6.8 ⌬prtV ⌬VC1649 show an almost-normal villous structure with minimum hemorrhage in the
mucosa, submucosa, and lamina propria; F, OMVs from the VC1649 complemented strain show accumulation of inflammatory cells and hemorrhage in mucosal
layers. (C) CT-beads ELISA results for OMVs from the C6709, CHA6.8, CHA6.8 ⌬prtV, CHA6.8⌬prtV ⌬VC1649, and Comp VC1649 strains. Culture super-
natant of strain N16961 was used as a positive control.
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1485Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
tease activity was detected in hapA and prtV mutant strains (10).
Sikora et al. reported the presence of several serine proteases, i.e.,
VesA, VesB, and VesC, in V. cholerae O1 El Tor strain N16961, in
which significantly less HAP is produced due to a frameshift mu-
tation in HapR (38). However, Altindis et al. showed the presence
of VesC in the OMVs of V. cholerae wild-type strain C6706 when
grown under conditions which activate TCP (22). HAP was not
detected in the OMVs of the wild-type strain under these condi-
tions, as production of HAP is coordinated in a fashion reciprocal
to that for cholera toxin and TCP (39). Secretion of HAP from
OMVs of V. cholerae is influenced by serine protease DegP, and
the degP mutant strains have been shown to secrete HAP and 8
other proteins from outer membrane vesicles (22).
Immunoblotting results showed the presence of mature 45-
kDa HAP in C6709 OMVs instead of its processed 35-kDa form
secreted through the TIISS. The mature 45-kDa form of HAP was
FIG 6 Comparative analysis of colonization of Vibrio cholerae strain C6709
and its isogenic knockout mutants in mouse intestine. A summary of the data,
represented as mean Ϯ SD (n ϭ 10 mice per group), is shown. Variables were
compared for significance using two-way analysis of variance and the Bonfer-
roni test (*, P value between 0.01 and 0.05; **, P value between 0.01 to 0.001).
FIG 5 Host cell death induced by OMVs from V. cholerae strains. (A) Flow cytometric analysis of cell death induced by V. cholerae OMVs. The graphical displays
show data from a representative experiment. In each display the lower right quadrant represents apoptotic cells (annexin Vϩ
/PIϪ
), and the upper left and right
quadrants represent necrotic cells (annexin Vϩ
/PIϩ
and annexin VϪ
/PIϩ
fractions). Upper panel, untreated Int407 cells and Int407 cells treated with 20 ␮g/ml
and 40 ␮g/ml of OMVs from V. cholerae strain C6709. Lower panel, Int407 cells treated with 20 ␮g/ml and 40 ␮g/ml OMVs from isogenic protease mutants of
the C6709 and Comp VC1649 strains. (B) Results from three independent flow cytometry experiments. Data are represented as mean (ϮSD) percentage of dead
cells after treatment with 20 and 40 ␮g/ml of OMVs from V. cholerae strains (n ϭ 3). (C) Fractions of apoptotic and necrotic cells of the same experiment. Data
are represented as mean (ϮSD) percentages of apoptotic and necrotic cells (n ϭ 3).
Mondal et al.
1486 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
purified earlier by Ghosh et al. in the presence of the metallopro-
tease inhibitor EDTA, whereas in its absence the processed 35-kDa
form is purified from culture supernatant (8). The protease un-
dergoes several steps of processing, including cleavage of the signal
peptide and further processing of the N terminus and C terminus
to generate the 35-kDa protein (4). The presence of the 45-kDa
form of HAP within the C6709 OMVs could be due to its lu-
minal localization within the vesicles. Earlier studies have
shown that luminal localization of proteins within OMVs pro-
tects them from proteolytic digestions (40). Therefore, it may
be suggested that the 45-kDa HAP is protected within the
OMVs from further processing.
OMVs have been shown to deliver different proteins and tox-
ins into host cells in an active form (40). Several studies have
shown that toxins such as CT, RTX, VCC, and metalloprotease
PrtV in V. cholerae are transported to host cells through OMVs in
a biologically active form (19, 20, 41). Our present study showed
that both HAP- and VesC-associated OMVs are internalized into
human intestinal epithelial cells (Int407) in a protease-indepen-
dent manner. Confocal microscopic images revealed that these
OMVs may be internalized into host cells through lipid rafts, as
vesicular endocytosis was completely inhibited by M␤CD and
nystatin.
The proteases in OMVs are transported in biologically active
forms. OMV-associated HAP induces dose-dependent morpho-
logical changes in Int407 cells similar to those shown by purified
HAP in our earlier study (8). HAP has been shown to proteolyti-
cally activate El Tor hemolysin/cytolysin by nicking at its N-ter-
minal peptide to generate the 65-kDa mature form (7). Our results
showed that OMV-associated HAP also digests the 79-kDa pro-
hemolysin to generate 65-kDa and 50-kDa mature forms. The
50-kDa mature hemolysin is a C-terminally processed, identically
truncated monomer of the 65-kDa mature hemolysin reported by
Ekigai et al. in V. cholerae O1 strains (42).
The roles of OMV-associated proteases in pathogenesis were
studied in the mouse ileal loop (MIL) model. This is the first study
to show fluid accumulation in mouse ileal loops caused by V.
cholerae OMV-associated proteases. HAP-associated OMVs
showed an enterotoxic response in the MIL assay. VesC-associated
CHA6.8 ⌬prtV OMVs showed a visible hemorrhagic fluid re-
sponse in MILs. Histopathological analysis of the mice ileum
showed grossly disrupted villi with hemorrhage in all layers of the
mucosa, which was significantly reduced upon treatment with
CHA6. 8⌬prtV ⌬VC1649 OMVs. These effects were restored with
OMVs from the VC1649 complemented strain. A similar hemor-
rhagic fluid response was observed with partially purified VesC
and culture supernatants from CHA6.8 ⌬prtV in rabbit ileal loop
(RIL) assay by Syngkon et al. They showed significant reduction of
hemorrhagic fluid when inhibited with PMSF (10). This hemor-
rhagic fluid accumulation was not due to cholera toxin, as CT-
FIG 7 IL-8 induction by OMVs of V. cholerae O1 strains in the T84 cell line. (A) Comparative analysis of IL-8 secretion from T84 cells coincubated with OMVs
from V. cholerae O1 El Tor strain C6709, its isogenic protease mutant strains CHA6.8, CHA6.8 ⌬prtV, and CHA6.8 ⌬prtV ⌬VC1649, and CHA6.8 ⌬prtV
⌬VC1649::VC1649 (Comp VC1649) determined by ELISA. Data represent mean Ϯ SD from three independent experiments performed under similar condi-
tions. Variables were compared for significance using two-way analysis of variance and the Bonferroni test (*, P value between 0.01 and0.05; **, P value between
0.01 and 0.001). (B) IL-8 production from T84 cells was analyzed at the mRNA and protein levels by RT-PCR and resolved in a 2% agarose gel. IL-8 expression
in T84 cells treated with OMVs from O1 El Tor strain C6709 and its protease knockout mutant strains CHA6.8, CHA6.8 ⌬prtV, CHA6.8 ⌬prtV ⌬VC1649, and
Comp VC1649 is displayed in lanes 2 to 5, respectively, and the untreated control is in lane 1. Densitometric quantification in densitometric units (DU) for IL-8
after normalization to GAPDH is shown below the agarose gel.
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1487Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
bead ELISA showed the absence of CT in V. cholerae OMVs (Fig.
4C). The OMV-free culture supernatants also showed the absence
of CT in bead ELISA (data not shown). This may be due to differ-
ent conditions for production of CT and HAP. Benitez et al.
showed that growth conditions that favor production of HAP
downregulate CT and TCP and vice versa (43).
OMV-associated proteases of V. cholerae induced cytotoxic ef-
fects in Int407 cells in a dose-dependent manner. C6709 OMVs
induced an apoptotic response in Int407 cells. Thay et al. have
shown that membrane-derived vesicles from Staphylococcus au-
reus induce apoptosis in a dose-dependent manner by an ␣-toxin-
dependent mechanism (29). However, the apoptotic cell popula-
tion was not significantly reduced when treated with CHA6.8
OMVs. CHA6.8 OMVs induced a greater cytotoxic response with
an increase in necrotic cells compared to HAP-associated C6709
OMVs. Studies have shown that there is an increased virulence in
hapA-deleted V. cholerae strains. Benitez et al. showed that inac-
tivation of hapA results in increased intestinal colonization and
adherence of V. cholerae in human intestinal epithelial cells (44).
Zhou et al. also reported that hapA-defective cholera vaccine can-
didate strains showed reactogenicity. Culture supernatants of
these strains can stimulate IL-8 secretion from T-84 cells (45).
Therefore, it may be suggested that the presence of metallopro-
tease PrtV in CHA6.8 OMVs may be responsible for increased
OMV-mediated cytotoxicity. However, earlier studies have shown
that PrtV modulates IL-8 secretion from T84 cells (46). Interest-
ingly, when cells were treated with CHA6.8 ⌬prtV, a shift from an
apoptotic population to a necrotic population was observed. In
CHA6.8 ⌬prtV ⌬VC1649 OMVs, the cytotoxicity was significantly
reduced, and increased cytotoxicity in Int407 cells was restored
with OMVs from the VC1649 complemented strain. These results
are very similar to the effect observed with OMVs on MIL assay.
Earlier studies on the pathogenicity of OMVs in bacterial in-
fections have shown the role of lipopolysaccharide (LPS)- and
OMV pathogen-associated molecular patterns (47, 48). LPS in the
OMVs is sensed by the Toll-like receptor 4 (TLR4) complex to
trigger inflammatory responses (49). OMVs isolated from Salmo-
nella enterica serovar Typhimurium activate macrophages and
dendritic cells to increase levels of surface major histocompatibil-
ity complex (MHC) class II expression as well as the production of
the proinflammatory mediators tumor necrosis factor alpha
(TNF-␣) and interleukin-12 (IL-12) (50). Helicobacter pylori and
Pseudomonas aeruginosa OMVs show a proinflammatory re-
sponse by IL-8 secretion (51, 52). IL-8 is a major proinflammatory
cytokine which act as a potent chemoattractant for polymorpho-
nuclear leukocytes (PMN) and recruits them into infected sites of
the epithelial layer, which results in opening of epithelial tight
junctions (53). The IL-8 response of Vibrio cholerae OMVs was
shown by Chatterjee and Chaudhuri. They showed that OMVs
from classical V. cholerae strain O395 induce IL-8 secretion from
intestinal epithelial cells, which activates dendritic cells to pro-
mote T-cell polarization (54). However, our results showed that
HAP-associated OMVs from El Tor strain C6709 failed to induce
significant IL-8 secretion from T84 cells. Earlier studies have also
shown that HAP does not induce IL-8 secretion from intestinal
epithelial cells (45). OMVs from ⌬hapA strain CHA6.8 showed
ϳ10-fold-increased IL-8 secretion compared to untreated con-
trols, whereas IL-8 secretion was increased ϳ25-fold upon treat-
ment with CHA6.8 ⌬prtV OMVs. The greater IL-8 response by
OMVs from the ⌬hapA ⌬prtV double mutant strain could be due
to the absence of PrtV, as it has been reported to reduce IL-8
secretion from intestinal epithelial cells (46). This increased IL-8
secretion by CHA6.8 ⌬prtV was significantly reduced upon treat-
ment with OMVs from the VesC mutant strain. Interestingly,
OMVs from Comp VC1649 showed significant induction of IL-8
secretion. These results show that VesC within the V. cholerae
OMVs is responsible for the increased IL-8 response in T84 cells.
Our results revealed 3-fold-increased colonization of the
CHA6.8 ⌬prtV strain compared to the VesC-deleted
CHA6.8⌬prtV⌬VC1649 strain in adult mice. Interestingly in-
creased colonization of V. cholerae was restored with the VC1649
complemented strain. Earlier, Silva et al. (59) also reported an
increased colonization of hapA mutant strains in infant mouse
model. However, Sikora et al. reported that VesC is not responsi-
ble for colonization of V. cholerae in infant mouse model (38). Our
results showed that VesC may play an important role in coloniza-
tion in adult mice.
OMVs have recently been used to develop acellular vaccines for
several diseases related to Gram-negative bacteria. The most suc-
cessful use of OMVs as a vaccine candidate has been against sero-
group B Neisseria meningitides infection, for which over 55 million
doses have been administered to date against serogroup B infec-
tions (55). Schild et al. showed that OMVs from V. cholerae can
generate a protective immune response against cholera infection
in the adult mouse model (25). However, our results suggest that
the presence of proteases in the OMVs may affect the use of OMVs
as a vaccine candidate for cholera.
In conclusion, we showed that the proteases HAP and VesC are
secreted in association with V. cholerae OMVs in an active form.
OMV-associated VesC induces a hemorrhagic response in the
MIL assay and a proinflammatory response in human cultured
intestinal epithelial cells, and VesC may play an important role in
colonization in the adult mouse model.
ACKNOWLEDGMENTS
This work was supported by intramural funding from the Indian Council
of Medical Research. A.M. was supported by a fellowship from the Indian
Council of Medical Research.
We thank Somnath Chatterjee, confocal microscope operator, NICED
central facility (Carl Zeiss, India), for technical help with confocal micro-
scopic studies, Somdatta Chatterjee, Division of Bacteriology, NICED, for
crucial assistance with VC1649 complementation, and Amarshi Mukher-
jee, Division of Biochemistry, NICED, for providing purified hemolysin.
REFERENCES
1. Farmer P, Almazor CP, Bahnsen ET, Barry D, Bazile J, Bloom BR, Bose
N, Brewer T, Calderwood SB, Clemens JD, Cravioto A, Eustache E,
Jérôme G, Gupta N, Harris JB, Hiatt HH, Holstein C, Hotez PJ, Ivers
LC, Kerry VB, Koenig SP, Larocque RC, Léandre F, Lambert W, Lyon
E, Mekalanos JJ, Mukherjee JS, Oswald C, Pape JW, Gretchko Prosper
A, Rabinovich R, Raymonville M, Réjouit JR, Ronan LJ, Rosenberg ML,
Ryan ET, Sachs JD, Sack DA, Surena C, Suri AA, Ternier R, Waldor
MK, Walton D, Weigel JL. 2011. Meeting cholera’s challenge to Haiti and
the world: a joint statement on cholera prevention and care. PLoS Negl
Trop Dis 5:e1145. http://dx.doi.org/10.1371/journal.pntd.0001145.
2. Faruque SM, Albert MJ, Mekalanos JJ. 1998. Epidemiology, genetics,
and ecology of toxigenic Vibrio cholerae. Microbiol Mol Biol Rev 62:1301–
1314.
3. Miyoshi S. 2013. Extracellular proteolytic enzymes produced by human
pathogenic Vibrio species. Front Microbiol 4:339. http://dx.doi.org/10
.3389/fmicb.2013.00339.
4. Häse CC, Finkelstein RA. 1991. Cloning and nucleotide sequence of the
Vibrio cholerae hemagglutinin/protease (HA/protease) gene and con-
struction of an HA/protease-negative strain. J Bacteriol 173:3311–3317.
Mondal et al.
1488 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
5. Finkelstein RA, Boesman-Finkelstein M, Chang Y, Häse CC. 1992.
Vibrio cholerae hemagglutinin/protease, colonial variation, virulence, and
detachment. Infect Immun 60:472–478.
6. Booth BA, Boesman-Finkelstein M, Finkelstein RA. 1984. Vibrio chol-
erae hemagglutinin/protease nicks cholera enterotoxin. Infect Immun 45:
558–560.
7. Nagamune K, Yamamoto K, Honda T. 1997. Intramolecular chaperone
activity of the pro-region of Vibrio cholerae El Tor cytolysin. J Biol Chem
272:1338–1343. http://dx.doi.org/10.1074/jbc.272.2.1338.
8. Ghosh A, Saha DR, Hoque KM, Asakuna M, Yamasaki S, Koley H, Das
SS, Chakrabarti MK, Pal A. 2006. Enterotoxigenicity of mature 45-
kilodalton and processed 35-kilodalton forms of hemagglutinin protease
purified from a cholera toxin gene-negative Vibrio cholerae non-O1, non-
O139 strain. Infect Immun 74:2937–2946. http://dx.doi.org/10.1128/IAI
.74.5.2937-2946.2006.
9. Vaitkevicius K, Lindmark B, Ou G, Song T, Toma C, Iwanaga M, Zhu
J, Andersson A, Hammarström ML, Tuck S, Wai SN. 2006. A Vibrio
cholerae protease needed for killing of Caenorhabditis elegans has a role in
protection from natural predator grazing. Proc Natl Acad Sci U S A 103:
9280–9285. http://dx.doi.org/10.1073/pnas.0601754103.
10. Syngkon A, Elluri S, Koley H, Rompikunata PK, Saha DR, Chakrabarti
MK, Bhadra RK, Wai SN, Pal A. 2010. Studies on a novel serine protease
of a ⌬hapA⌬prtV Vibrio cholerae O1 strain and its role in haemorrhagic
response in the rabbit ileal loop model. PLoS One 5:e13122. http://dx.doi
.org/10.1371/journal.pone.0013122.
11. Pugsley AP. 1993. The complete general secretory pathway in gram-
negative bacteria. Microbiol Rev 57:50–108.
12. Kuehn MJ, Kesty NC. 2005. Bacterial outer membrane vesicles and host
pathogen interaction. Genes Dev 19:2645–2655. http://dx.doi.org/10
.1101/gad.1299905.
13. Wai SN, Lindmark B, Soderblom T, Takade A, Westermark M, Os-
carsson J, Jass J, Richter-Dhalfors A, Mizunoe Y, Uhlin BE. 2003.
Vesicle-mediated export and assembly of pore-forming oligomers of the
enterobacterial ClyA cytotoxin. Cell 115:25–35. http://dx.doi.org/10.1016
/S0092-8674(03)00754-2.
14. Balsalobre C, Silvan JM, Berglund S, Mizunoe Y, Uhlin BE, Wai SN.
2006. Release of the type I secreted alpha-haemolysin via outer membrane
vesicles from Escherichia coli. Mol Microbiol 59:99–112. http://dx.doi.org
/10.1111/j.1365-2958.2005.04938.x.
15. Davis JM, Carvalho HM, Rasmussen SB, O’Brien AD. 2006. Cytotoxic
necrotizing factor type 1 delivered by outer membrane vesicles of uro-
pathogenic E. coli attenuates polymorphonuclear leukocyte antimicrobial
activity and chemotaxis. Infect Immun 74:4401–4408. http://dx.doi.org
/10.1128/IAI.00637-06.
16. Lindmark B, Rompikuntal PK, Vaitkevicius K, Song T, Mizunoe Y,
Uhlin BE, Guerry P, Wai SN. 2009. Outer membrane vesicle-mediated
release of cytolethal distending toxin (CDT) from Campylobacter jejuni.
BMC Microbiol 9:220. http://dx.doi.org/10.1186/1471-2180-9-220.
17. Parker H, Chitcholtan K, Hampton MB, Keenan JI. 2010. Uptake of
Helicobacter pylori outer membrane vesicles by gastric epithelial cells. In-
fect Immun 78:5054–5061. http://dx.doi.org/10.1128/IAI.00299-10.
18. Bomberger JM, MacEachran DP, Coutermarsh BA, Ye S, O’Toole GA,
Stanton BA. 2009. Long-distance delivery of bacterial virulence factors by
Pseudomonas aeruginosa outer membrane vesicles. PLoS Pathog
5:e1000382. http://dx.doi.org/10.1371/journal.ppat.1000382.
19. Chatterjee D, Chaudhuri K. 2011. Association of cholera toxin with
Vibrio cholerae outer membrane vesicles which are internalized by human
intestinal epithelial cells. FEBS Lett 585:1357–1362. http://dx.doi.org/10
.1016/j.febslet.2011.04.017.
20. Elluri S, Enow C, Vdovikova S, Rompikunal P, Dongre M, Carlsson S,
Pal A, Uhlin BE, Wai SN. 2014. Outer membrane vesicles mediate
transport of biologically active Vibrio cholerae cytolysin (VCC) from V.
cholerae strains. PLoS One 9:e106731. http://dx.doi.org/10.1371/journal
.pone.0106731.
21. Rompikuntal PK, Vdovikova S, Duperthuy M, Jhonson TL, Ahlund M,
Lundmark R, Oscarson J, Sandkvist M, Uhlin BE, Wai SN. 2015. Outer
membrane vesicle mediated export of processed PrtV protease from
Vibrio cholerae. PLoS One 10:e0134098. http://dx.doi.org/10.1371/journal
.pone.0134098.
22. Altindis E, Fu Y, Mekalanos JJ. 2014. Proteomic analysis of Vibrio chol-
erae outer membrane vesicles. Proc Natl Acad Sci U S A 111:E1548–
E1556. http://dx.doi.org/10.1073/pnas.1403683111.
23. Gosink KK, Kobayashi R, Kawagishi I, Hase CC. 2002. Analyses of the
roles of three cheA homologs in chemotaxis of Vibrio cholerae. J Bacteriol
184:1767–1771. http://dx.doi.org/10.1128/JB.184.6.1767-1771.2002.
24. Chen DJ, Osterrieder N, Metzger SM, Buckles E, Doody AM, DeLisa
MP, Putnam D. 2010. Delivery of foreign antigens by engineered outer
membrane vesicle vaccines. Proc Natl Acad Sci U S A 107:3099–3104.
http://dx.doi.org/10.1073/pnas.0805532107.
25. Schild S, Nelson EJ, Camilli A. 2008. Immunization with Vibrio cholerae
outer membrane vesicles induces protective immunity in mice. Infect Im-
mun 76:4554–4563. http://dx.doi.org/10.1128/IAI.00532-08.
26. Cheng LW, Schneewind O. 2000. Yersinia enterocolitica TyeA, an intra-
cellular regulator of the type III machinery, is required for specific target-
ing of YopE, YopH, YopM, and YopN into the cytosol of eukaryotic cells.
J Bacteriol 182:3183–3190. http://dx.doi.org/10.1128/JB.182.11.3183
-3190.2000.
27. Parker H, Chitcholtan K, Hampton MB, Keenan JI. 2010. Uptake of
Helicobacter pylori outer membrane vesicles by gastric epithelial cells. In-
fect Immun 72:5054–5061. http://dx.doi.org/10.1128/IAI.00299-10.
28. Sawasvirojwong S, Srimanote P, Chatsudthipong V, Muanprasat C.
2013. An adult mouse model of Vibrio cholerae induced diarrhea for study-
ing pathogenesis and potential therapy of cholera. PLoS Negl Trop Dis
7:e2293. http://dx.doi.org/10.1371/journal.pntd.0002293.
29. Thay B, Wai SN, Oscarsson J. 2013. Staphylococcus aureus ␣-toxin de-
pendent induction of host cell death by membrane derived vesicles. PLoS
One 8:e54661. http://dx.doi.org/10.1371/journal.pone.0054661.
30. Oku Y, Uesaka Y, Hirayama T, Takeda Y. 1988. Development of highly
sensitive bead-ELISA to detect bacterial protein toxins. Microbiol Immu-
nol 32:807–816. http://dx.doi.org/10.1111/j.1348-0421.1988.tb01442.x.
31. Olivier V, Queen J, Satchell KJF. 2009. Successful small intestine colo-
nization of adult mice by Vibrio cholerae requires ketamine anesthesia and
accessory toxins. PLoS One 4:e7352. http://dx.doi.org/10.1371/journal
.pone.0007352.
32. Chatterjee SN, Das J. 1967. Electron microscopic observations on the
excretion of cell-wall material by Vibrio cholerae. J Gen Microbiol 49:1–11.
http://dx.doi.org/10.1099/00221287-49-1-1.
33. Rodal SK, Skretting G, Garred F, Vilhardt B, Van Duers B, Sandvig.
1999. Extraction of cholesterol with methyl-beta-cyclodextrin perturbs
formation of clathrin-coated endocytic vesicles. Mol Biol Cell 10:961–974.
http://dx.doi.org/10.1091/mbc.10.4.961.
34. Bonnington KE, Kuehn MJ. 2014. Protein secretion and export via outer
membrane vesicles. Biochim Biophys Acta 1843:1612–1619. http://dx.doi
.org/10.1016/j.bbamcr.2013.12.011.
35. Sheikh IA, Koley H, Chakrabarti MK, Hoque KM. 2013. The Epac
signalling pathway regulates ClϪ
secretion via modulation of apical
KCNN4c channels in diarrhea. J Biol Chem 288:20404–20415. http://dx
.doi.org/10.1074/jbc.M113.467860.
36. Scott ME, Dossani ZY, Sandkvist M. 2001. Directed polar secretion of
protease from single cells of Vibrio cholerae via the type II secretion path-
way. Proc Natl Acad Sci U S A 98:13978–13983. http://dx.doi.org/10.1073
/pnas.241411198.
37. Young DB, Broadbent DA. 1982. Biochemical characterization of extra-
cellular proteases from Vibrio cholerae. Infect Immun 37:875–883.
38. Sikora AE, Zielke RA, Lawrence DA, Andrews PC, Sandkvist M. 2011.
Proteomic analysis of the Vibrio cholerae type II secretome reveals new
proteins, including three related serine proteases. J Biol Chem 286:16555–
16566. http://dx.doi.org/10.1074/jbc.M110.211078.
39. Silva AJ, Pham K, Benitez JA. 2003. Hemaglutinnin/protease expression
and mucin gel penetration in El Tor biotype Vibrio cholerae. Microbiology
149:1883–1891. http://dx.doi.org/10.1099/mic.0.26086-0.
40. Kulp A, Kuehn MJ. 2010. Biological functions and biogenesis of secreted
bacterial outer membrane vesicles. Annu Rev Microbiol 64:163–184. http:
//dx.doi.org/10.1146/annurev.micro.091208.073413.
41. Boardman BK, Meehan BM, Fullner-Satchell KJ. 2007. Growth phase
regulation of Vibrio cholerae RTX toxin export. J Bacteriol 189:1827–1835.
http://dx.doi.org/10.1128/JB.01766-06.
42. Ikigai H, Ono T, Nakae T, Otsuru H, Shimamura T. 1999. Two form of
Vibrio cholerae O1 El Tor hemolysin derived from identical precursor
protein. Biochim Biophys Acta 1415:297–305. http://dx.doi.org/10.1016
/S0005-2736(98)00183-7.
43. Benitez JA, Silva AJ, Finkelstein RA. 2001. Environmental signals con-
trolling production of hemagglutinin/protease in Vibrio cholerae. Infect
Immun 69:6549–6553. http://dx.doi.org/10.1128/IAI.69.10.6549-6553
.2001.
44. Bénitez JA, Spelbrink RG, Silva A, Phillips TE, Stanley CM, Boesman-
OMV-Associated Active Proteases in V. cholerae
May 2016 Volume 84 Number 5 iai.asm.org 1489Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
Finkelstein M, Finkelstein RA. 1997. Adherence of Vibrio cholerae to
cultured differentiated human intestinal cells: an in vitro colonization
model. Infect Immun 65:3474–3477.
45. Zhou X, Gao DQ, Michalski J, Benitez JA, Kaper JB. 2004. Induction of
interleukin-8 in T84 cells by Vibrio cholerae. Infect Immun 72:389–397.
http://dx.doi.org/10.1128/IAI.72.1.389-397.2004.
46. Ou G, Rompikuntal PK, Bitar A, Lindmark B, Vaitkevicius K, Wai SN,
Hammarström ML. 2009. Vibrio cholerae cytolysin causes an inflamma-
tory response in human intestinal epithelial cells that is modulated by the
PrtV protease. PLoS One 4:e7806. http://dx.doi.org/10.1371/journal.pone
.0007806.
47. Whitmire WM, Garon CF. 1993. Specific and nonspecific responses of
murine B cells to membrane blebs of Borrelia burgdoferi. Infect Immun
61:1460–1467.
48. Opal SM. 2007. The host response to endotoxin, antilipopolysaccaride
strategies, and the management of severe sepsis. Int J Med Microbiol 297:
365–377. http://dx.doi.org/10.1016/j.ijmm.2007.03.006.
49. Ellis TN, Kuehn MJ. 2010. Virulence and immunomodulatory roles of
bacterial outer membrane vesicles. Microbiol Mol Biol Rev 74:81–94. http:
//dx.doi.org/10.1128/MMBR.00031-09.
50. Alaniz RC, Deatherage BL, Lara JC, Cookson BT. 2007. Membrane
vesicles are immunogenic facsimiles of Salmonella typhimurium that po-
tently activate dendritic cells, prime B and T cell responses, and stimulate
protective immunity in vivo. J Immunol 179:7692–7701. http://dx.doi.org
/10.4049/jimmunol.179.11.7692.
51. Bauman SJ, Kuehn MJ. 2006. Purification of outer membrane vesicles
from Pseudomonas aeruginosa and their activation of an IL-8 response.
Microbes Infect 8:2400–2408. http://dx.doi.org/10.1016/j.micinf.2006.05
.001.
52. Ismail S, Hampton MB, Keenan JI. 2003. Helicobacter pylori outer mem-
brane vesicles modulate proliferation and interleukin-8 production by
gastric epithelial cells. Infect Immun 71:5670–5675. http://dx.doi.org/10
.1128/IAI.71.10.5670-5675.2003.
53. McCormick BA, Siber AM, Maurelli AT. 1998. Requirement of the
Shigella flexneri virulence plasmid in the ability to induce trafficking of
neutrophils across polarized monolayers of the intestinal epithelium. In-
fect Immun 66:4237–4243.
54. Chatterjee D, Chaudhuri K. 2013. Vibrio cholera O395 outer membrane
vesicles modulate intestinal epithelial cells in a NOD1 dependent manner
and induces dendritic cell-mediated Th2/Th17 responses. J Biol Chem
288:4299–4309. http://dx.doi.org/10.1074/jbc.M112.408302.
55. Holst J, Martin D, Arnold R, Huergo CC, Oster P, O’Hallahhan J,
Rosenqvist E. 2009. Properties and clinical performance of vaccines con-
taining outer membrane vesicles from Neisseria meningitides. Vaccine 27:
B3–B12. http://dx.doi.org/10.1016/j.vaccine.2009.04.071.
56. Haralalka S, Nandi S, Bhadra RK. 2003. Mutation in the relA gene of
Vibrio cholerae affects in vitro and in vivo expression of virulence factors.
J Bacteriol 185:4672–4682. http://dx.doi.org/10.1128/JB.185.16.4672
-4682.2003.
57. Jung HC, Eckmann L, Yang SK, Panja A, Fierer J, Morzycka-
Wroblewska E, Kagnoff MF. 1995. A distinct array of proinflammatory
cytokines is expressed in human colon epithelial cells in response to bac-
terial invasion. J Clin Invest 95:55–65. http://dx.doi.org/10.1172
/JCI117676.
58. Sarkar M, Chaudhuri K. 2004. Association of adherence and motility in
interleukin 8 induction in human intestinal epithelial cells by Vibrio chol-
erae. Microbes Infect 6:676–685. http://dx.doi.org/10.1016/j.micinf.2004
.02.018.
59. Silva AJ, Leitch GJ, Camilli A, Benitez JA. 2006. Contribution of hem-
agglutinin/protease and motility to the pathogenesis of El Tor biotype
cholera. Infect Immun 74:2072–2079. http://dx.doi.org/10.1128/IAI.74.4
.2072-2079.2006.
Mondal et al.
1490 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity
onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom

More Related Content

What's hot

2010Ketorolac
2010Ketorolac2010Ketorolac
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
Eneutron
 
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
Farhad B. Hashemi, PhD
 
J World's Poult Res 5(2) 21-28, 2015
J World's Poult Res 5(2) 21-28, 2015J World's Poult Res 5(2) 21-28, 2015
J World's Poult Res 5(2) 21-28, 2015
Hany Ellakany
 
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
Tiffany Chen
 
Antituberculosis and toxicity assay of ethanolic extract of mimba cortex
Antituberculosis and toxicity assay of ethanolic extract of mimba cortexAntituberculosis and toxicity assay of ethanolic extract of mimba cortex
Antituberculosis and toxicity assay of ethanolic extract of mimba cortex
Cut Fatimah
 
My CV_final
My CV_finalMy CV_final
My CV_final
Arif Jamal
 
A cost effective scheme developed for studying human malaria caused by plasmo...
A cost effective scheme developed for studying human malaria caused by plasmo...A cost effective scheme developed for studying human malaria caused by plasmo...
A cost effective scheme developed for studying human malaria caused by plasmo...
Alexander Decker
 
Nehad m. sayed (ain shams)1
Nehad m. sayed (ain shams)1Nehad m. sayed (ain shams)1
Nehad m. sayed (ain shams)1
yousef2009
 
AMR Collaborative Grants at the University of Bristol
AMR Collaborative Grants at the University of BristolAMR Collaborative Grants at the University of Bristol
AMR Collaborative Grants at the University of Bristol
warwick_amr
 
Thesis Defense Presentation
Thesis Defense PresentationThesis Defense Presentation
Thesis Defense Presentation
Stephanie Libby, M.S.
 
Ab
AbAb
Elucidating the role of the Chromosomal Type III Secretion System structural ...
Elucidating the role of the Chromosomal Type III Secretion System structural ...Elucidating the role of the Chromosomal Type III Secretion System structural ...
Elucidating the role of the Chromosomal Type III Secretion System structural ...
Jackson Osaghae-Nosa
 
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indicaAntiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
Alexander Decker
 
MaximoGuerrieri_Bio_Mac
MaximoGuerrieri_Bio_MacMaximoGuerrieri_Bio_Mac
MaximoGuerrieri_Bio_Mac
Maximo Guerrieri
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
inventionjournals
 
Univ bacteriav PCR Primer
Univ bacteriav PCR PrimerUniv bacteriav PCR Primer
Univ bacteriav PCR Primer
Zulkifli As
 
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
Santhi Devasundaram
 
Nuhu et al_Poster NAPA2016 correction and observation
Nuhu et al_Poster NAPA2016 correction and observationNuhu et al_Poster NAPA2016 correction and observation
Nuhu et al_Poster NAPA2016 correction and observation
Nuhu Tanko
 
J0361058062
J0361058062J0361058062
J0361058062
inventionjournals
 

What's hot (20)

2010Ketorolac
2010Ketorolac2010Ketorolac
2010Ketorolac
 
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
Specific prophylaxis & therapy of infectious diseases. Immune sera & Immunogl...
 
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
Activation-of-human-immunodeficiency-virus-type-1-expression-by-Gardnerella-v...
 
J World's Poult Res 5(2) 21-28, 2015
J World's Poult Res 5(2) 21-28, 2015J World's Poult Res 5(2) 21-28, 2015
J World's Poult Res 5(2) 21-28, 2015
 
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
Daily Intake of Probiotics with High IFN-γ IL-10 Ratio Increases the Cytotoxi...
 
Antituberculosis and toxicity assay of ethanolic extract of mimba cortex
Antituberculosis and toxicity assay of ethanolic extract of mimba cortexAntituberculosis and toxicity assay of ethanolic extract of mimba cortex
Antituberculosis and toxicity assay of ethanolic extract of mimba cortex
 
My CV_final
My CV_finalMy CV_final
My CV_final
 
A cost effective scheme developed for studying human malaria caused by plasmo...
A cost effective scheme developed for studying human malaria caused by plasmo...A cost effective scheme developed for studying human malaria caused by plasmo...
A cost effective scheme developed for studying human malaria caused by plasmo...
 
Nehad m. sayed (ain shams)1
Nehad m. sayed (ain shams)1Nehad m. sayed (ain shams)1
Nehad m. sayed (ain shams)1
 
AMR Collaborative Grants at the University of Bristol
AMR Collaborative Grants at the University of BristolAMR Collaborative Grants at the University of Bristol
AMR Collaborative Grants at the University of Bristol
 
Thesis Defense Presentation
Thesis Defense PresentationThesis Defense Presentation
Thesis Defense Presentation
 
Ab
AbAb
Ab
 
Elucidating the role of the Chromosomal Type III Secretion System structural ...
Elucidating the role of the Chromosomal Type III Secretion System structural ...Elucidating the role of the Chromosomal Type III Secretion System structural ...
Elucidating the role of the Chromosomal Type III Secretion System structural ...
 
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indicaAntiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
Antiplasmodial efficacy of fruit extracts and cladodes of opuntia ficus indica
 
MaximoGuerrieri_Bio_Mac
MaximoGuerrieri_Bio_MacMaximoGuerrieri_Bio_Mac
MaximoGuerrieri_Bio_Mac
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
 
Univ bacteriav PCR Primer
Univ bacteriav PCR PrimerUniv bacteriav PCR Primer
Univ bacteriav PCR Primer
 
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
T cell recall response of two hypothetical proteins (Rv2251 and Rv2721c) from...
 
Nuhu et al_Poster NAPA2016 correction and observation
Nuhu et al_Poster NAPA2016 correction and observationNuhu et al_Poster NAPA2016 correction and observation
Nuhu et al_Poster NAPA2016 correction and observation
 
J0361058062
J0361058062J0361058062
J0361058062
 

Viewers also liked

V. cholerae
V. choleraeV. cholerae
V. cholerae
Samer Bio
 
Identification of vibrio cholerae pathogenicity
Identification of vibrio cholerae pathogenicityIdentification of vibrio cholerae pathogenicity
Identification of vibrio cholerae pathogenicity
Maharaj Vinayak Global University
 
Vibrio cholerae
Vibrio choleraeVibrio cholerae
Vibrio cholerae
camiij1
 
2 pp vibrio
2 pp vibrio2 pp vibrio
2 pp vibrio
Bruno Mmassy
 
Vibrio cholera
Vibrio choleraVibrio cholera
Vibrio cholera
Hassan Qureshi
 
Vibrio cholerae
Vibrio choleraeVibrio cholerae
Vibrio cholerae
Kanikanigoro
 
Vibrio Colera
Vibrio ColeraVibrio Colera
Cholera powerpoint
Cholera powerpointCholera powerpoint
Cholera powerpoint
Kari900
 

Viewers also liked (8)

V. cholerae
V. choleraeV. cholerae
V. cholerae
 
Identification of vibrio cholerae pathogenicity
Identification of vibrio cholerae pathogenicityIdentification of vibrio cholerae pathogenicity
Identification of vibrio cholerae pathogenicity
 
Vibrio cholerae
Vibrio choleraeVibrio cholerae
Vibrio cholerae
 
2 pp vibrio
2 pp vibrio2 pp vibrio
2 pp vibrio
 
Vibrio cholera
Vibrio choleraVibrio cholera
Vibrio cholera
 
Vibrio cholerae
Vibrio choleraeVibrio cholerae
Vibrio cholerae
 
Vibrio Colera
Vibrio ColeraVibrio Colera
Vibrio Colera
 
Cholera powerpoint
Cholera powerpointCholera powerpoint
Cholera powerpoint
 

Similar to PAPER1

The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
Pauline Ogrodzki
 
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficileThe HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
Joost Verhoeks
 
Respiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestineRespiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestine
Andrew Fabich
 
Kit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile DuctKit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile Duct
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDFPREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
Nuhu Tanko
 
1ScIeNtIFIc REPORTS (2018) 81250 DOI10.1038s41598-018.docx
1ScIeNtIFIc REPORTS   (2018) 81250   DOI10.1038s41598-018.docx1ScIeNtIFIc REPORTS   (2018) 81250   DOI10.1038s41598-018.docx
1ScIeNtIFIc REPORTS (2018) 81250 DOI10.1038s41598-018.docx
vickeryr87
 
ajcmi-02-03-29691 edited by SC
ajcmi-02-03-29691 edited by SCajcmi-02-03-29691 edited by SC
ajcmi-02-03-29691 edited by SC
KOBRA SALIMIYAN rizi
 
viruses-08-00300 (1)
viruses-08-00300 (1)viruses-08-00300 (1)
viruses-08-00300 (1)
Fabrizio Di Pinto
 
Agro[2]
Agro[2]Agro[2]
Agro[2]
Dilip Pandya
 
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
Marios Stylianou
 
Muthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journalMuthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journal
DrMuni Neurophysiologist
 
bacteriospermia
bacteriospermiabacteriospermia
PIIS2210909915000661
PIIS2210909915000661PIIS2210909915000661
PIIS2210909915000661
Mohsen Tabasi
 
Lactonase RSC Printed copy
Lactonase RSC Printed copyLactonase RSC Printed copy
Lactonase RSC Printed copy
Chetan Meena
 
IJAA
IJAAIJAA
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
iosrjce
 
21022_ftp
21022_ftp21022_ftp
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
Mariela Marín
 
OBASM-fd
OBASM-fdOBASM-fd
OBASM-fd
Cory Kozlovich
 
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Mohamed Khalid Ali Xundhur
 

Similar to PAPER1 (20)

The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
 
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficileThe HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
The HtrA-Like Protease CD3284 Modulates Virulence of Clostridium difficile
 
Respiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestineRespiration of e. coli in the mouse intestine
Respiration of e. coli in the mouse intestine
 
Kit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile DuctKit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile Duct
 
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDFPREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
PREVALENCE AND ANTIMICROBIAL SUSCEPTIBILITY OF ESBL IN SOKOTO PDF
 
1ScIeNtIFIc REPORTS (2018) 81250 DOI10.1038s41598-018.docx
1ScIeNtIFIc REPORTS   (2018) 81250   DOI10.1038s41598-018.docx1ScIeNtIFIc REPORTS   (2018) 81250   DOI10.1038s41598-018.docx
1ScIeNtIFIc REPORTS (2018) 81250 DOI10.1038s41598-018.docx
 
ajcmi-02-03-29691 edited by SC
ajcmi-02-03-29691 edited by SCajcmi-02-03-29691 edited by SC
ajcmi-02-03-29691 edited by SC
 
viruses-08-00300 (1)
viruses-08-00300 (1)viruses-08-00300 (1)
viruses-08-00300 (1)
 
Agro[2]
Agro[2]Agro[2]
Agro[2]
 
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
Marios Stylianou_Paper II_ Novel High-Throughput Screening Method for Identif...
 
Muthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journalMuthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journal
 
bacteriospermia
bacteriospermiabacteriospermia
bacteriospermia
 
PIIS2210909915000661
PIIS2210909915000661PIIS2210909915000661
PIIS2210909915000661
 
Lactonase RSC Printed copy
Lactonase RSC Printed copyLactonase RSC Printed copy
Lactonase RSC Printed copy
 
IJAA
IJAAIJAA
IJAA
 
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
Effect of pH concentration on hydatid cyst protoscolices infectivity: In vitr...
 
21022_ftp
21022_ftp21022_ftp
21022_ftp
 
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
Leishmania amazonensis INFECTION INDUCES CHANGES IN POTASSIUM PERMEABILITY OF...
 
OBASM-fd
OBASM-fdOBASM-fd
OBASM-fd
 
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
 

PAPER1

  • 1. Cytotoxic and Inflammatory Responses Induced by Outer Membrane Vesicle-Associated Biologically Active Proteases from Vibrio cholerae Ayan Mondal,a Rima Tapader,a Nabendu Sekhar Chatterjee,b Amit Ghosh,a * Ritam Sinha,c Hemanta Koley,c Dhira Rani Saha,d Manoj K. Chakrabarti,a Sun Nyunt Wai,e Amit Pala Division of Pathophysiology,a Division of Biochemistry,b Division of Bacteriology,c and Histology and Electron Microscopy,d National Institute of Cholera and Enteric Diseases, Kolkata, India; Department of Molecular Biology, Umea Centre of Microbial Research, Umea University, Umea, Swedene Proteases in Vibrio cholerae have been shown to play a role in its pathogenesis. V. cholerae secretes Zn-dependent hemaggluti- nin protease (HAP) and calcium-dependent trypsin-like serine protease (VesC) by using the type II secretion system (TIISS). Our present studies demonstrated that these proteases are also secreted in association with outer membrane vesicles (OMVs) and transported to human intestinal epithelial cells in an active form. OMV-associated HAP induces dose-dependent apoptosis in Int407 cells and an enterotoxic response in the mouse ileal loop (MIL) assay, whereas OMV-associated VesC showed a hemor- rhagic fluid response in the MIL assay, necrosis in Int407 cells, and an increased interleukin-8 (IL-8) response in T84 cells, which were significantly reduced in OMVs from VesC mutant strain. Our results also showed that serine protease VesC plays a role in intestinal colonization of V. cholerae strains in adult mice. In conclusion, our study shows that V. cholerae OMVs secrete biolog- ically active proteases which may play a role in cytotoxic and inflammatory responses. Vibrio cholerae is the causative agent of the life-threatening dis- ease cholera. Cholera epidemics in Haiti in 2010 provide evi- dence that it still remains an ongoing public health threat (1). Strains of the El Tor biotype O1 serogroup are responsible for seventh pandemic and recent cholera outbreaks (2). Cholera toxin (CT) and toxin-coregulated pilus (TCP) have been identified as the major virulence factors for V. cholerae pathogenesis. CT is responsible for profuse watery diarrhea, whereas TCP is essential for sustaining colonization of human small intestine. V. cholerae also secretes several proteases which may also play a role in its pathogenesis (3). The major protease secreted by V. cholerae strains is the 35-kDa hemagglutinin protease (HAP) (4). HAP has been reported to accelerate the bacterial detachment from cultured cells by diges- tion of V. cholerae adhesins (5). HAP modulates the enterotoxige- nicity of cholera toxin by nicking the A subunit of CT (6). HAP also plays a role in processing hemolysin to its mature form by removal of a 15-kDa N-terminal peptide (7). As shown in the above-mentioned studies, HAP plays an indirect role in V. chol- erae pathogenesis. The possibility of its direct role in pathogenesis has been shown by Ghosh et al. (8). They reported that purified HAP from a V. cholerae non-O1, non-O139 strain showed a hem- orrhagic response in rabbit ileal loops (RILs) and an increase in intestinal short-circuit current in Ussing’s chamber (8). Besides HAP, the other major well-characterized metalloprotease is a 97- kDa Vibrio cholerae protease, PrtV which has been shown to play a role in virulence in the Caenorhabditis elegans infection model (9). In an earlier study, we also reported the presence of a novel serine protease encoded by VC1649 (VesC) in a hapA prtV mutant Vibrio cholerae O1 strain and showed its role in the hemorrhagic re- sponse in the rabbit ileal loop model (10). Gram-negative bacteria, including V. cholerae, use 6 different secretion systems (type I to type VI) to transport important viru- lence factors to the cell envelope and the extracellular milieu (11). Outer membrane vesicles (OMVs) may also serve as a mechanism of delivering active virulence factors into host cells. The virulence factors are protected from digestion by host proteases and are secreted not individually but as multiple factors delivered in a discrete package (12). Toxin delivery mediated by OMVs is recog- nized as a potent virulence mechanism for many bacterial patho- gens. Association of toxins with OMVs have been reported for many pathogenic Gram-negative bacteria. Toxins such as ClyA cytotoxin, ␣-hemolysin, and CNF1 of Escherichia coli, cytolethal distending toxin (CDT) of Campylobacter jejuni, and vacuolating cytotoxin A (VacA) of Helicobacter pylori are secreted through OMVs (13, 14, 15, 16, 17). Bomberger et al. showed that multiple virulence factors, such as ␤-lactamase, alkaline phosphatase, he- molytic phospholipase C, and Cif, are directly delivered into the host cytoplasm via fusion of OMVs with lipid rafts in the host plasma membrane and that these toxins are rapidly distributed to specific subcellular locations to affect host cell biology (18). OMV-mediated secretion of biologically active CT, Vibrio chol- erae cytolysin (VCC), and PrtV has been reported in V. cholerae (19, 20, 21). Recent studies on proteomic analysis of V. cholerae OMVs have the shown presence of both HAP and the 59-kDa serine protease VesC (22). However, the functions of these vesicle-associated pro- teases have not been elucidated. Our study shows that V. cholerae OMVs secrete biologically active proteases which may play a role in cytotoxic and inflammatory responses. Received 5 November 2015 Returned for modification 23 December 2015 Accepted 20 February 2016 Accepted manuscript posted online 29 February 2016 Citation Mondal A, Tapader R, Chatterjee NS, Ghosh A, Sinha R, Koley H, Saha DR, Chakrabarti MK, Wai SN, Pal A. 2016. Cytotoxic and inflammatory responses induced by outer membrane vesicle-associated biologically active proteases from Vibrio cholerae. Infect Immun 84:1478–1490. doi:10.1128/IAI.01365-15. Editor: A. Camilli Address correspondence to Amit Pal, apal7364@rediffmail.com. * Present address: Amit Ghosh, Department of Physiology, All India Institute of Medical Sciences, Bhubaneswar, India. Copyright © 2016, American Society for Microbiology. All Rights Reserved. crossmark 1478 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 2. MATERIALS AND METHODS Ethics statement. Animal experiments were performed after obtaining necessary permission from the Institutional Animal Ethical Committee (IAEC). The IAEC/CPCSEA approval number is APRO 7624/11/2010. Bacterial strains. The bacterial strains, and oligonucleotides used in this study are listed in Table 1. V. cholerae cells were routinely grown in tryptic soy broth (TSB) (Difco, USA) at 37°C with shaking as described earlier (10). E. coli TOP 10 cells were grown overnight in Luria broth (LB) at 37°C with shaking at 200 rpm. Antibiotics were used at the following concentrations: streptomycin, 100 ␮g/ml; kanamycin, 40 ␮g/ml; and am- picillin, 100 ␮g/ml. Bacterial cells, including mutants, were maintained at Ϫ70°C in LB containing 20% sterile glycerol. Complementation of the VC1649 gene in V. cholerae strain CHA6.8 ⌬prtV ⌬VC1649. The VC1649 gene was amplified from genomic DNA isolated from CHA6.8 ⌬prtV using VesC forward and reverse primers (Table 1). Cloning was performed as described by Gosink et al. (23). The amplified VC1649 gene was cloned into the pBAD-TOPO TA (Invitrogen, USA) cloning vector under the control of an arabinose-inducible pro- moter using a cloning kit from Invitrogen and was transformed in E. coli TOP 10 cells. Transformed colonies (Ampϩ ) were selected in a Luria agar (LA) plate containing 100 ␮g/ml ampicillin. Positive clones were selected from the transformed colonies by colony PCR with the pBAD forward and VesC reverse primers supplied in the cloning kit, and plasmids were iso- lated using a plasmid isolation kit (Thermo Fisher, USA). Plasmid pVC1649 was transformed into V. cholerae strain CHA6.8 ⌬prtV ⌬VC1649 by electroporation (23). IsolationandpurificationofOMVsfromV.choleraestrains.V.chol- erae cells were grown in 2 liter of TSB for 18 h at 37°C under shaking conditions, and outer membrane vesicles (OMVs) were prepared from late-log-phase culture as described previously (17). Crude vesicles were isolated from strain Comp VC1649 grown in 250 ml of TSB using L-ara- binose induction as described by Chen et al. (24). Isolated crude OMVs from C6709, OMVs from its isogenic protease mutants, and OMVs from Comp VC1649 were purified by density gradient centrifugation on a dis- continuous Optiprep-iodixanol (Sigma, USA) gradient as described by Elluri et al. (20). Fractions of equal volumes were sequentially removed after density gradient centrifugation and analyzed by Western blotting using anti-OmpU antibody. OmpU is the marker protein for V. cholerae OMVs. The protein content of purified OMVs was estimated using the Bradford assay. Transmission electron microscopy (TEM). Twenty micrograms of purified OMVs was placed on carbon-coated nickel grids (300 mesh) (Sigma, USA), stained with 2% aqueous uranyl acetate, rinsed, and exam- ined with a transmission electron microscope operating at 60 kV (FEI Tecnai 12; Bio, Twin, The Netherlands). RaisingofantiseraagainstHAP,VesC,andOMVs.HAPwaspurified from the culture supernatant of C6709 as described by Ghosh et al. (8). Antiserum to purified HAP was prepared by immunizing a New Zealand White rabbit by intramuscular injection with 100 ␮g of HAP emulsified with an equal volume of Freund’s complete adjuvant (Sigma, USA). This was followed by four booster injections with 100 ␮g of HAP and incom- plete adjuvant (Sigma, USA) at 7-day intervals. Blood samples were col- lected from rabbits on day 0 and 3 days after the final injections and were allowed to clot at room temperature for 30 min. Sera were collected, followed by centrifugation (1,000 rpm, 10 min), dilution in phosphate- buffered saline (PBS) (Sigma, USA), and storage at Ϫ80°C (25). Antise- rum against the 59-kDa serine protease (VesC) was also raised in a New Zealand White rabbit. VesC was partially purified as described earlier (10). The mass spectrometrically identified band of VesC in a native poly- acrylamide gel was excised, homogenized in PBS, and administered into an intramuscular region of a New Zealand White rabbit. Antiserum against VesC was drawn from the rabbit after four booster injections as described above. Antiserum against OMVs was prepared by immunizing 7-week-old mice (BALB/c). Mice were immunized subcutaneously with 50 ␮g of OMVs isolated from C6709 at the first, second, and third weeks, followed by a booster dose of 75 ␮g of the OMVs at the fourth week. The serum was collected, and the antibody titer was determined by enzyme- linked immunosorbent assay (ELISA) with the preimmune serum as a control. SDS-PAGE and Western blot analysis. V. cholerae OMVs were sepa- rated by 10% SDS-PAGE. Briefly, 20 ␮g each of purified OMVs and whole-cell lysates of C6709 and CHA6.8 were mixed with Laemmli sample TABLE 1 Bacterial strains, plasmids, and primers Strain, plasmid, or primer Relevant characteristics Source or reference Vibrio cholerae strains C6709 Wild type (O1 El Tor); Smr 56 CHA6.8 C6709 ⌬hapA::Kan; Smr Kmr 10 CHA6.8 ⌬prtV CHA6.8 ⌬prtV; Smr Kmr 10 CHA6.8 ⌬prtV ⌬VC1649 (VesC mutant) CHA6.8 ⌬prtV ⌬VC1649; Smr Kmr 10 Comp VC1649 CHA6.8 ⌬prtV ⌬VC1649::pVC1649; Smr Kmr Ampr This study E. coli strain TOP 10 FϪ mcrA ⌬(mrr-hsdRMS-mcrBC) ␾80lacZ⌬M15⌬lacX74 recA1 araD139 ⌬(ara-leu)7697 galU galK rpsL (Strr ) endA1 nupG Invitrogen, USA Plasmids pBAD TOPO-TA Ampϩ TA cloning vector plasmid Invitrogen, USA pVC1649 pBAD TOPO with Vibrio cholerae VC1649 (VesC) This study Primers IL 8 F1 5=-ATGGGGAAGGTGAAGGTCGG-3= 57 IL 8 R1 5=-TCTCAGCCCTCTTCAAAAACTTCTC-3= 57 GAPDH F1 5=-ATGGGGAAGGTGAAGGTCGG-3= 58 GAPDH R1 5=-GGATGCTAAGCAGTTGGT-3= 58 pBAD F 5=-ATGCCATAGCATTTTTATCC-3= Invitrogen, USA pBAD R 5=-GATTTAATCTGTATCAGG-3= Invitrogen, USA VesC F 5=-ATGAACAAAACATTTTTATC-3= This study VesC R 5=-TCAGACGCGTTGACGACG-3= This study OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1479Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 3. buffer (1:1) and boiled for 10 min prior to electrophoresis. Proteins with known molecular weights (Thermo Fisher, USA) were used as markers. Gels were stained with Coomassie brilliant blue stain after electrophoresis. For immunoblotting, the separated proteins were transferred onto a ni- trocellulose membrane (Bio-Rad, USA) by the wet transfer method using a Trans-Blot apparatus (Bio-Rad, USA). The membrane was blocked at room temperature for 1 h in 5% nonfat dry milk powder (Bio-Rad, USA)– PBS–Tween 20. After incubation, the membrane was washed and incu- bated with rabbit anti-HAP (1:1,000 dilution in 5% [wt/vol] nonfat dry milk), and anti-VesC (1:10,000 dilution in 5% [wt/vol] nonfat dry milk) antisera, followed by incubation for 1 h at room temperature with a 1:10,000 dilution of anti-rabbit horseradish peroxidase-conjugated sec- ondary antibody (Thermo Fisher, USA). The blots were visualized with ECL Western blotting detection reagent (Millipore, USA). Proteolytic digestion of prohemolysin. Proteolytic digestion of El Tor prohemolysin was performed as described earlier (7). Briefly, 10 ␮l of 79-kDa prohemolysin (1 mg/ml) was mixed with 10 ␮l (100 ␮g/ml) of purified C6709 OMVs and incubated at 37°C for 30 min, and SDS-PAGE was performed. Untreated purified prohemolysin and HAP-treated pro- hemolysin were used as negative and positive controls, respectively. Proteinase K susceptibility assay. The proteinase K susceptibility as- say was performed as described previously (26). Briefly, OMVs were treated with proteinase K (1 ␮g/ml) for 30 min at 37°C in either the presence or absence of 1% SDS, and samples were analyzed by immuno- blotting against anti-HAP and anti-VesC antibodies raised in rabbit. Media for cell lines and culture conditions. Int407 and T84 human intestinal epithelial cells were used in this study. Int407 cells were grown in 25-ml tissue culture flasks (BD Biosciences) containing minimal essential medium (MEM) (Gibco, USA) supplemented with 10% fetal bovine se- rum (Gibco, USA) along with penicillin G and streptomycin sulfate (Sigma, USA) at 37°C in a CO2 incubator (Heraeus, Germany). Cells at 80 to 90% confluence were seeded in 6-well and 24-well tissue culture plates (BD Biosciences) at a concentration of approximately 105 cells/well. Cells were permitted to reach confluence by allowing them to grow for 24 h in a CO2 incubator. T84 cells were used for interleukin-8 (IL-8) assay, and the cells were maintained in Dulbecco’s modified Eagle’s Medium-nutri- ent F12 (DMEM-F12) (Gibco, USA) supplemented with antibiotic solu- tion as described above. Tissue culture assay. The tissue culture assay was performed as de- scribed by Ghosh et al. (8). Briefly, OMVs from C6709 and its isogenic knockout strains were filter sterilized and serially diluted in serum-free MEM from 0.5 ␮g/ml to 100 ␮g/ml. The exhaust media were aspirated from 24-well tissue culture plates containing Int407 cells grown to con- fluence and washed with PBS, and serially diluted OMVs were added and grown at 37°C in a CO2 incubator for 24 h. Purified HAP was used as positive control. Results were analyzed by phase-contrast microscopy and photographs taken (CK 40; Olympus). OMV internalization assay. Int407 cells were incubated with 20 ␮g/ml of purified OMVs from C6709 and its isogenic protease mutants for 1 h at 37°C in a 5% CO2 atmospheric chamber to monitor internaliza- tion of V. cholerae OMVs. The effects of several endocytic inhibitors (nys- tatin, dynasore, and methyl-␤-cyclodextrin [M␤CD]) on OMV uptake were examined by pretreating Int407 cells with nystatin (25 ␮g/ml) and dynasore (80 ␮M) for 1 h and with M␤CD (1 mg/ml) (Sigma, USA) for 30 min as described by Parker et al. (27). This was followed by incubation with purified OMVs for 1 h. Cells were then fixed with 4% paraformalde- hyde at 4°C for 20 min, followed by permeabilization with 1% Triton X-100 in PBS for 20 min at room temperature. Mouse antiserum against OMVs (1:2,000) was added to OMV-treated cells and incubated overnight at 4°C in a moist chamber. Cells were washed with sterile PBS and incu- bated with tetramethyl rhodamine isocyanate (TRITC)-conjugated anti- mouse secondary antibody (1:500) and were kept in the dark at 37°C for 90 min. For colocalization studies, primary antibodies against HAP (1:1,000) and OMVs (1:2,000) raised in rabbit and mouse, respectively, were added to C6709 and CHA6.8 OMV-treated cells. Primary antibodies against VesC (1:2,500) and OMVs (1:2,000) were added to cells treated with OMVs from C6709, isogenic protease mutants of C6709, and the VC1649 complemented strain and incubated overnight as described above. Cells were incubated with fluorescein isothiocyanate (FITC)-conjugated anti- rabbit IgG (1:500) and TRITC-conjugated anti-mouse antibody (1:500) and were kept in the dark at 37°C for 90 min after incubation. Cells were washed and resuspended in 100 ␮l of PBS. Approximately 20 ␮l of resus- pended sample was used to make a thin film on a glass slide. One drop of 90% glycerol was added, a thin glass coverslip was placed on it, and the slide was viewed under a confocal microscope (model LSM 510 META; Zeiss, Germany) and excited at 488 nm and 543 nm for FITC and TRITC, respectively. The emission filters used were 475 nm to 525 nm for FITC and 558 nm to 601 nm for TRITC. Experiments were also done with PBS as negative control. Merged images were developed using Image J soft- ware. Mouse intestinal fluid accumulation assay. The mouse intestinal fluid accumulation assay was performed as described earlier (28). Briefly, 20 ␮g of purified OMVs from V. cholerae and its isogenic protease mutant strains was injected into ligated intestinal segments of about 4 cm in length from 4- to 5-week-old adult BALB/c mice. All experimental proce- dures were performed under anesthesia with ketamine. After 6 h, mice were sacrificed and loops were excised. The enterotoxic activity was de- termined by measuring the fluid accumulation (FA) ratio, which is the ratio of fluid accumulated in the intestinal loop to the length of the loop (g/cm). PBS was used as a negative control. Values were expressed as mean Ϯ standard deviation (SD) of the mean (n ϭ 5 mice per group). Histopathological studies of mouse ileal loops (MILs). Mouse ileal tissues were collected and fixed in 10% neutral buffered formalin solution for histopathological studies. The tissues were further embedded in par- affin and processed following the standard protocol. Three- to four-mi- crometer thin sections were cut in a microtomy rotor (Leica, Germany). The sections were strained with hematoxylin and eosin and observed un- der a light microscope. Photographs were taken under a magnification of ϫ40 with a Leica DMLB microscope (Solms, Germany) equipped with a digital imaging system. Flow cytometric analysis. Flow cytometric analysis was performed as described by Thay et al. (29). Briefly, Int 407 cells were treated with OMVs from V. cholerae strain C6709 and its protease knockout mutant strains at concentrations of 20 ␮g/ml and 40 ␮g/ml for 4 h and then stained with fluorescein isothiocyanate (FITC)-conjugated annexin V and propidium iodide (PI) (BD Pharmingen) according to the manufacturer’s instruc- tions. The cells were stained with FITC-annexin V in annexin V binding buffer for 10 min, followed by addition of PI, and analyzed by flow cy- tometry using Cell Quest Pro software (BD Biosciences). A total of 104 cells were acquired for data analysis in each set of experiments. Values were expressed as mean Ϯ SD of the mean for 3 different observations (n ϭ 3). CT-bead ELISA. CT concentrations in OMVs and culture superna- tants of C6709 and its isogenic protease mutants grown in TSB were mea- sured by bead ELISA as described by Oku et al. (30). Culture supernatant of V. cholerae N16961 grown in Syncase medium was used as a positive control. Mouse intestinal colonization assay. Pathogen-free adult 4- to 5-week-old BALB/c mice were used for the colonization assay. Prior to the experiment, all mice were fasted for 24 h with free access to sterile water. The experiment was performed as described by Olivier et al. (31). Briefly, mice were injected intraperitoneally with a mixture of 35 mg/kg ketamine and 5 mg/kg xylazine for anesthesia. Two hundred microliters of suspen- sions of ϳ109 viable bacterial cells was administered orally with two bo- luses of 0.5 ml of 5% (wt/vol) NaHCO3. All mice were kept in microiso- lator cages with free access to food and sterile water after inoculation. Mice were euthanized with Euthanasia 6 solution after 18 h of oral inoculation. The small intestine above the cecum was collected after dissection, ho- mogenized, and then serially diluted in sterile PBS (pH 7.4). Homoge- Mondal et al. 1480 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 4. nized materials were plated on appropriate antibiotic-containing tryptic soy agar (TSA) plates and incubated for 24 h. All experiments were per- formed in triplicates, and values were expressed as mean Ϯ SD (n ϭ 10 mice per group) (*, P Ͻ 0.05; **, P Ͻ 0.01) and compared with the CFU obtained for C6709. ELISA and semiquantitative RT-PCR for IL-8 estimation. For en- zyme-linked immunosorbent assays (ELISAs), the growth medium was removed from the plates. Cells were washed with PBS (Sigma, USA) and maintained in antibiotic- and serum-free DMEM-F12. The cells were treated with 10 ␮g/ml of V. cholerae OMVs from C6709 and its isogenic knockout mutant strains for IL-8 stimulation. Supernatants were col- lected after 24 h and centrifuged to remove dead cells, and IL-8 concen- trations were determined by ELISA using an ELISA kit from Invitrogen (USA) according to the instructions of the manufacturer. For reverse transcription-PCR (RT-PCR), total RNA was isolated from the OMV- treated T84 cells using TRIzol reagent (Invitrogen, USA). RNA was quan- tified by spectrophotometric analysis and the quality of the RNA samples determined by estimating the A260/A280 ratio. cDNA was prepared from 1.5 ␮g of total RNA using the Superscript II first-strand synthesis system for RT-PCR (Invitrogen, USA). A 2.5-␮l portion of cDNA was PCR am- plified in a 25-␮l reaction volume containing 10 mM Tris-Cl (pH 8.3), 50 mM KCl, 2.0 mM MgCl2, 2.5 mM each deoxynucleoside triphosphate (dNTP), 20 pmol of each primer, and 1 U of Taq DNA polymerase (Roche, Germany). The primers used for IL-8 and GAPDH (glyceralde- hyde-3-phosphate dehydrogenase) are listed in Table 1. The PCR condi- tions were as follows: an initial denaturation step at 95°C for 5 min fol- lowed by 35 cycles of 95°C (1 min), 55°C and 60°C for IL-8 and GAPDH, respectively (1 min), and 72°C (30 s). The final extension step was done at 72°C for 7 min. The products were electrophoresed on a 2% agarose gel, stained with ethidium bromide, and visualized using a gel documentation system (Bio-Rad, USA). The data from ELISA and RT-PCR was recorded as mean Ϯ SD from at least three groups of experiments. Densitometric quantification of RT-PCR bands was performed with Quantity One soft- ware (Bio-Rad, USA). Statistical analysis. All experiments were performed in triplicates. Values were expressed as mean Ϯ standard deviation of the mean. Vari- ables were compared for significance using two-way analysis of variance and the Bonferroni test, with one asterisk indicating a P value between 0.01 to 0.05 and two asterisks indicating a P value between 0.01 to 0.001. RESULTS Fractions of HAP and VesC are associated with OMVs of Vibrio cholerae C6709 and hapA mutant strains, respectively. V. chol- erae secretes abundant vesicles during its normal course of growth (32). To investigate the association of proteases with outer mem- brane vesicles (OMVs), crude OMVs were isolated from culture supernatants of the pathogenic O1 El Tor strain C6709 and its protease mutants by ultracentrifugation, followed by purification using Optiprep density gradient centrifugation. Fractions ob- tained from the gradient centrifugation of OMVs were analyzed by immunoblotting using OmpU antiserum. Fractions showing reactivity with OmpU antiserum were examined by transmission electron microscopy for ultrastructural analysis and used for fur- ther experiments. OMVs of C6709 appeared as spherical electron- dense materials of heterogeneous sizes with a diameter in the range of 25 to 150 nm. Major morphological differences were not observed in OMVs from the isogenic protease mutant strains and OMVs from Comp VC1649. To investigate OMV-associated se- cretion of HAP, OMVs from HAP-producing C6709 and ⌬hapA strain CHA6.8 were subjected to SDS-PAGE along with whole-cell lysates from the respective strains, and immunoblotting was per- formed using polyclonal antiserum against HAP. Concentrated OMV-free culture supernatants from C6709 and purified HAP were used as positive controls. An immunoreactive band corre- sponding to HAP was observed only in C6709 OMVs. The pres- ence of HAP was not observed in whole-cell lysate prepared from strain C6709. The immunoblot results showed the presence of the mature 45-kDa form of HAP in OMVs instead of its processed 35-kDa form, which was present in OMV-free culture supernatant of C6709 (Fig. 1B). The presence of VesC in the OMVs was de- tected by immunoblotting using VesC antiserum (1:10,000). VesC was detected in the OMVs from the CHA6.8 and CHA6.8 ⌬prtV strains and the VC1649 complemented construct. In contrast, its presence was not observed in OMVs from C6709 and CHA6.8 ⌬prtV ⌬VC1649 (Fig. 1C). The above results demonstrated that FIG 1 Association of HAP and VesC with V. cholerae OMVs by immunoblot analysis. (A) Ten percent SDS-PAGE profile of the OMVs of Vibrio cholerae strains C6709 (lane 3), CHA6.8 (lane 4), CHA6.8 ⌬prtV (lane 5), CHA6.8 ⌬prtV ⌬VC1649 (lane 6), and Comp VC1649 (complemented) (lane 7). Five micrograms of purified HAP was resolved in lane 2, and a prestained protein ladder (Thermo Fisher, USA) is in lane 1. (B) Immunoblot analyses with OMVs from C6709 and CHA6.8 and whole-cell lysates from the respective strains against HAP antiserum. Purified 45-kDa and 35-kDa HAP and concentrated OMV-free culture supernatant of C6709 were used as positive controls. (C) Immunoblot analyses with OMVs of C6709 and its isogenic protease mutants against VesC antiserum. OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1481Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 5. HAP is secreted in association with OMVs from the V. cholerae wild-type strain C6709, whereas the serine protease VesC is se- creted in association with OMVs from ⌬hapA strains. LocalizationofproteasewithintheOMVs.Localizationofthe HAP and VesC within the OMVs was confirmed by proteinase K susceptibility assay. Clear proteolytic digestion of HAP was ob- served only when OMVs from C6709 were incubated with protei- nase K in the presence of 1% SDS, and this proteolytic digestion was inhibited in the presence of phenylmethylsulfonyl fluoride (PMSF) (Fig. 2A). A similar result was observed with OMV-asso- ciated VesC in the CHA6.8 ⌬prtV strain (Fig. 2B). These results confirmed the luminal localization of HAP and VesC within the vesicles. Protease-associated OMVs bind to and are internalized by human intestinal epithelial cells in a protease-independent manner. The interaction of OMVs with Int407 cells was studied by confocal microscopy. OMV-specific red fluorescence was ob- served throughout Int407 cells treated with OMVs from C6709 and its isogenic protease mutant strains (Fig. 3A, panels 4 and 7, and C, panels 1, 4, 7, and 10). In contrast, no fluorescence was observed in cells treated with buffer (Fig. 3A, panel 1). Internal- ization of OMVs in Int407 cells was confirmed by treatment with different endocytic inhibitors prior to addition of OMVs. The cholesterol-sequestering agent M␤CD inhibited the entry of V. cholerae OMVs in Int407 cells (Fig. 3B, panels 5 and 6). This result suggests that V. cholerae OMVs may be internalized into the cells by cholesterol-enriched lipid rafts. M␤CD has also been reported to inhibit clathrin-mediated endocytosis when applied at high concentrations (33). Inhibition of vesicular endocytosis was fur- ther confirmed by using nystatin, a cholesterol binding agent, which disrupts lipid rafts but does not affect clathrin-mediated endocytosis. Our results showed that nystatin also inhibits the entry of V. cholerae OMVs in Int407 cells (Fig. 3B, panels 7 and 8). Interestingly, dynasore (an inhibitor of the dynamin-dependent endocytic pathway) failed to inhibit internalization of OMVs into Int407 cells (Fig. 3B, panels 9 and 10). The inhibition of internal- ization of C6709 OMVs is shown in Fig. 3B, which shows that C6709 OMVs failed to enter Int407 cells upon treatment with M␤CD and nystatin. The above results suggested that V. cholerae OMVs are internalized into In407 cells by lipid raft-dependent endocytosis. To investigate OMV-mediated transport of HAP in Int407 cells, confocal microscopy was performed, using HAP antiserum and FITC-conjugated secondary antibody. The merged images showed colocalization of HAP and OMVs in Int407 cells when treated with HAP-associated C6709 OMVs (Fig. 3A, panel 6), whereas cells treated with CHA6.8 showed only OMV-specific red fluorescence (Fig. 3A, panel 9). OMVs were labeled with VesC antiserum and FITC-conjugated secondary antibody to assess whether VesC is transported into cells. Our results with the colo- calization assay clearly showed that VesC is transported into Int407 cells through OMVs from hapA mutant strains CHA6.8 and CHA6.8 ⌬prtV (Fig. 3C, panels 6 and 9). Colocalization of OMVs and VesC was not observed when cells were treated with CHA6.8 ⌬prtV ⌬VC1649 OMVs. However, the merged image showed adherence and internalization of OMVs in the absence of VesC (Fig. 3C, panel 12), and this suggests that VesC is not respon- sible for adherence of OMVs in Int407 cells. OMV-mediated transport of VesC was further confirmed with OMVs from strain Comp VC1649, which showed both OMV- and VesC-specific sig- nals in the cells (Fig. 3C, panels 13 and 14). OMV-associated proteases are biologically active. Studies have shown that OMVs deliver cargo to the specific targets in an active form (34). In the present study, we showed that both HAP and VesC are transported into intestinal epithelial cells through OMVs of their producing strains. HAP-associated C6709 OMVs induced morphological changes of Int407 cells in a dose-depen- dent manner when treated for 24 h (Fig. 4A, panel I). C6709 OMVs showed a cell-distending effect at a low concentration (20 ␮g/ml), whereas they showed a cell-rounding effect similar to that for purified HAP at a higher concentration (50 ␮g/ml) (Fig. 4A, panel I). In contrast, such morphological changes were not ob- served when cells were treated with OMVs from CHA6.8 and other protease mutant strains. The proteolytic activity of HAP- associated OMVs was studied on 79-kDa prohemolysin to con- firm its biological activity. HAP has been reported to proteolyti- cally cleave prohemolysin by digestion of the N-terminal polypeptide chain to its 65-kDa mature form (7). Our results showed that OMV-associated HAP digests 79-kDa prohemolysin to produce 65-kDa and 50-kDa fragments, similar to the effect observed with purified HAP (Fig. 4A, panel II). Our results con- firmed that OMV-associated HAP is biologically active. In earlier studies, we have reported that purified HAP and partially purified VesC induce a hemorrhagic fluid response in the RIL assay (8, 10). The biological activities of OMV-associated proteases were evalu- ated in vivo by fluid accumulation assay in the ilea of adult mice. OMVs from wild-type strain C6709 showed significant fluid ac- cumulation (FA ratio, 0.20 Ϯ 0.03; n ϭ 5) (**, P Յ 0.01) compared to the control (PBS treated) (Fig. 4B, panel I). Histopathological studies of ileal tissues revealed widely dilated and ruptured villi and the presence of inflammatory cells in the mucosa (magnifica- tion, ϫ40) (Fig. 4B, panel IIB). Interestingly, hemorrhagic fluid accumulation was observed in the intestinal segments when they were treated with OMVs from CHA6.8 and CHA6.8 ⌬prtV. His- topathological studies of the ileal tissues showed an altered villous structure and mild hemorrhage in the submucosal membrane when treated with CHA6.8 OMVs (Fig. 4B, panel IIC). However, CHA6.8 ⌬prtV OMVs caused maximum damage of the ileal tis- sues (Fig. 4B, panel IID). The ileal tissues showed grossly dis- FIG 2 Localization of proteases in OMVs by proteinase K susceptibility assay. OMVs from C6709 and CHA6.8 ⌬prtV were incubated at 37°C for 30 min with or without 1 ␮g/ml proteinase K alone or with 1% SDS or 1 mM PMSF. Lane 1, control reaction; lane 2, sensitivity to extracellular protease; lane 3, inhibi- tion of extracellular proteases; lane 4, protease sensitivity to luminal proteins; lane 5, inhibition of protease activity by PMSF (control). OMVs were incu- bated under the above-described conditions, precipitated with acetone, dried, electrophoresed, and immunoblotted against HAP (A) or VesC (B) antiserum. Mondal et al. 1482 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 6. FIG 3 Colocalization of proteases and OMVs in the Int407 cell line. (A) Int407 cells were treated with OMVs from strain C6709 (panels 4, 5, and 6) or CHA6.8 (panels 7, 8, and 9) or with sterile PBS as a negative control (panels 1, 2, and 3), incubated with HAP and OMV antisera, and stained with TRITC (red)-conjugated anti-mouse antibody (panels 1, 4, and 7). Red fluorescence was detected in all OMVs except the negative control. HAP was detected using anti-HAP and FITC-conjugated (green) anti-rabbit antibody in panel 5. Merged images are shown in panels 3, 6, and 9. The nucleus was stained with DAPI (4=,6=-diamidino- 2-phenylindole). (B) Inhibition of internalization of C6709 OMVs in Int407 cells treated with dynasore, methyl-␤-cyclodextrin (M␤CD), and nystatin prior to OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1483Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 7. rupted villi with hemorrhage in all layers of the mucosa. In con- trast, ileal tissues treated with CHA6.8 ⌬prtV ⌬VC1649 OMVs showed an almost normal villous structure with minimum hem- orrhage in the mucosa and submucosa in histopathological stud- ies (Fig. 4B, panel IIE). Fluid accumulation in the intestinal seg- ments was also significantly reduced (FA ratio, 0.07 Ϯ 0.03; n ϭ 5). These results may suggest that OMV-associated VesC may be responsible for the hemorrhagic fluid response in the mouse ileal loop assay. The MIL assay with OMVs from Comp VC1649 showed restoration of hemorrhagic fluid in intestinal segments, and histopathological studies showed mucosal damage and the presence of inflammatory cells in the mucosa and submucosa (Fig. 4B, panel IIF). The presence of VesC in the OMVs may responsible for the hemorrhagic fluid responses. However, it was reported earlier that CT is responsible for fluid accumulation in the mouse ileal loop assay (35). We measured the CT titers of V. cholerae OMVs and culture supernatants by CT-bead ELISA to confirm whether the OMV-mediated inflammatory responses is due to the presence of CT in the OMVs. Both cell-free culture supernatants and OMVs from V. cholerae strains grown in TSB showed almost negligible CT titers compared to that of the positive-control strain N16961 grown in Syncase medium (Fig. 4C) Therefore, the hem- orrhagic response by OMVs in the MIL assay is not due to CT. The above results confirmed that OMV-associated VesC is responsible for hemorrhagic fluid accumulation in mouse ileal loops. Host cell death induced by V. cholerae OMVs is dependent on proteases. The cell death mechanisms induced by V. cholerae OMVs were analyzed by flow cytometry. Flow cytometric analysis with C6709 OMVs showed a dose-dependent apoptotic response in Int407 cells (annexin Vϩ /PIϪ fraction). More than 50% cell death was observed at 20 ␮g/ml of C6709 OMVs compared to the untreated control. However, CHA6.8 OMVs induced greater cy- totoxicity than C6709 OMVs, and cell death occurred by both apoptosis and necrosis. Interestingly 30% reduced cell death was observed when cell were treated with CHA6.8 ⌬prtV OMVs com- pared to CHA6.8 OMVs. However, a 2-fold increase in the ne- crotic cell population was observed compared to that with CHA6.8 OMVs (Fig. 5C). CHA6.8 ⌬prtV ⌬VC1649 OMVs showed reduced cytotoxicity compared to C6709, CHA6.8, and CHA6.8 ⌬prtV OMVs. OMVs from the VC1649 complemented strain restored the cytotoxicity in Int407 cells. These results clearly demonstrated that VesC is responsible for necrotic damage of Int407 cells (Fig. 5B and C). VesC plays a role in intestinal colonization of V. cholerae in the adult mouse model. Assessment of colonization of V. cholerae strains in mouse intestinal epithelium showed significantly re- duced bacterial colonization of the triple knockout mutant strain CHA6.8 ⌬prtV ⌬VC1649 (CFU 3 ϫ 103 cells/g of intestine) com- pared with the ⌬hapA ⌬prtV strain CHA6.8 ⌬prtV (CFU 8 ϫ 105 cells/g of intestine) (Fig. 6). The ⌬hapA strain CHA6.8 showed increased colonization (CFU 1 ϫ 108 cells/g of intestine) com- pared to wild-type strain C6709 (CFU 3 ϫ 107 cells/g of intestine). There was a significant decrease in colonization in the CHA6.8 ⌬prtV ⌬VC1649 strain compared to the CHA6.8 ⌬prtV strain. Interestingly, when the VC1649 gene was complemented in CHA6.8 ⌬prtV ⌬VC1649, the strain showed 3-fold-increased col- onization. These results suggest that VesC may play a role in col- onization in adult mice. OMV-mediated IL-8 secretion from T84 cells is dependent on VesC. OMVs isolated from C6709 and its protease mutant Vibrio cholerae strains were incubated with T84 cells for 24 h, and culture supernatants were taken and assayed for IL-8 expression by ELISA and semiquantitative RT-PCR. Significant IL-8 mRNA expression was observed when cells were treated with CHA6.8, CHA6.8 ⌬prtV, and Comp VC1649, as visible IL-8-specific bands were observed in agarose gel electrophoresis. OMVs from the CHA6.8 ⌬prtV and Comp VC1649 strains induced maximum mRNA expression (Fig. 7B). Protein expression in the same set of experiments was studied by IL-8 ELISA to assess whether the IL-8 mRNA level is correlated at the protein level. The ELISA results showed that OMVs from CHA6.8 induced more than a 2.5-fold increase in IL-8 secretion compared to those from wild-type strain C6709 and a ϳ10-fold increase compared to those from untreated cells. Interestingly, IL-8 secretion was increased ϳ25-fold com- pared to that in untreated cells when cells were treated with CHA6.8 ⌬prtV OMVs in 24 h. This increased IL-8 secretion was reduced 4-fold when cells were treated with OMVs from the VesC mutant strain (Fig. 7A). Upon VesC complementation in the hapA prtV VC1649 triple knockout mutant strain, IL-8 induction by OMVs was significantly increased. These results suggest that VesC may act as an inducer for OMV-mediated IL-8 secretion in human intestinal epithelial cells. DISCUSSION Proteases produced by microorganisms play an important role in virulence (3). Vibrio cholerae secretes hemagglutinin protease (HAP), V. cholerae protease (PrtV), and serine proteases through the type II secretion system (TIISS) (21, 36). Earlier we have re- ported that purified HAP from a ctx-negative V. cholerae non-O1, non-O139 strain and VesC from a ⌬hapA ⌬prtV Vibrio cholerae strain may play a role in pathogenesis by inducing a hemorrhagic response in the rabbit ileal loop (RIL) assay (8, 10). Our present study demonstrated that HAP and VesC are also secreted in asso- ciation with outer membrane vesicles (OMVs). Our results showed that HAP is secreted in association with OMVs from wild- type strain C6709, whereas the 59-kDa serine protease VesC is secreted in association with OMVs from hapA mutant strains but not from wild-type strain C6709. The absence of VesC in C6709 OMVs could be due to its proteolytic degradation in the presence of HAP, as it has been reported to mask the secretion of other proteases from V. cholerae (3, 10). Earlier studies have shown that secretion of serine proteases is regulated by metalloproteases in Vibrio species (3, 37). Syngkon et al. showed that 90% of the pro- tease activity in strain C6709 is due to HAP, as only residual pro- addition of OMVs. Cells with untreated OMVs were used as a negative control and cells without inhibition as a positive control. OMVs were labeled with TRITC-conjugated antibody, and the nucleus was stained with DAPI. (C) Int407 cells were treated with C6709 OMVs (panels 1, 2, and 3), CHA6.8 OMVs (panels 4, 5, and 6), CHA6.8 ⌬prtV OMVs (panels 7, 8, and 9), CHA6.8 ⌬prtV⌬ VC1649 OMVs (panels 10, 11, and 12), and Comp VC1649 (panels 13, 14, and 15), incubated with VesC and OMVs antisera, and stained with red TRITC-conjugated anti-mouse antibody (panels 1, 4, 7, 10, and 13). Red fluorescence were detected in all OMVs. The presence of VesC was detected using anti-VesC and FITC-conjugated (green) anti-rabbit antibodies (panels 5, 8, and 14). Merged images are shown in panels 3, 6, 9, 12, and 15. The nucleus was stained with DAPI. Mondal et al. 1484 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 8. FIG 4 Protease-associated OMVs are biologically active. (A) Biological activities of HAP-associated OMVs. Panel I, Int407 cells were treated with OMVs from C6709 at 50 ␮g/ml and 20 ␮g/ml and OMVs from CHA6.8 for 24 h at 37°C. The higher concentration of C6709 OMVs showed a cell-rounding effect, whereas the lower concentration showed a cell-distending effect. No morphological changes were observed in CHA6.8. Panel II, immunoblot analyses against hemolysin antiserum with 10 ␮g purified hemolysin treated with HAP and C6709 OMVs and purified hemolysin as positive control. (B) Panel I, effect of OMVs of Vibrio cholerae strains in the mouse ileal loop assay. Twenty micrograms (100 ␮g/ml) of OMVs from wild-type strain C6709 and its knockout derivatives was introduced into ligated ileal loops of adult mice. The mice were sacrificed after 6 h, loops were excised, and fluid accumulation was calculated as the loop weight/length ratio. Images of mouse ileal loops treated with V. cholerae OMVs are shown. The graph shows a summary of the data, represented as mean Ϯ SD (n ϭ 5 mice per group). Variables were compared for significance using two-way analysis of variance and the Bonferroni test (*, P value between 0.01 and 0.05; **, P value between 0.01 and 0.001). Panel II, histopathological studies of mouse ileal loops. Twenty micrograms of OMV-treated ileal tissues was processed for histopathological analysis, and photomicrographs were taken at a magnification of ϫ40. A, PBS-treated ileal tissues showed normal villi with mucosal structure; B, a magnified image of ileal tissues treated with C6709 OMVs shows dilated and ruptured villi with accumulation of inflammatory cells in mucosal layers; C, CHA6.8 shows an altered villous structure with mild hemorrhage in the submucosa; D, CHA 6.8 ⌬prtV OMV-treated ileal tissues show grossly disrupted villi with hemorrhage in all layers of the mucosa; E, ileal tissues treated with OMVs of strain CHA6.8 ⌬prtV ⌬VC1649 show an almost-normal villous structure with minimum hemorrhage in the mucosa, submucosa, and lamina propria; F, OMVs from the VC1649 complemented strain show accumulation of inflammatory cells and hemorrhage in mucosal layers. (C) CT-beads ELISA results for OMVs from the C6709, CHA6.8, CHA6.8 ⌬prtV, CHA6.8⌬prtV ⌬VC1649, and Comp VC1649 strains. Culture super- natant of strain N16961 was used as a positive control. OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1485Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 9. tease activity was detected in hapA and prtV mutant strains (10). Sikora et al. reported the presence of several serine proteases, i.e., VesA, VesB, and VesC, in V. cholerae O1 El Tor strain N16961, in which significantly less HAP is produced due to a frameshift mu- tation in HapR (38). However, Altindis et al. showed the presence of VesC in the OMVs of V. cholerae wild-type strain C6706 when grown under conditions which activate TCP (22). HAP was not detected in the OMVs of the wild-type strain under these condi- tions, as production of HAP is coordinated in a fashion reciprocal to that for cholera toxin and TCP (39). Secretion of HAP from OMVs of V. cholerae is influenced by serine protease DegP, and the degP mutant strains have been shown to secrete HAP and 8 other proteins from outer membrane vesicles (22). Immunoblotting results showed the presence of mature 45- kDa HAP in C6709 OMVs instead of its processed 35-kDa form secreted through the TIISS. The mature 45-kDa form of HAP was FIG 6 Comparative analysis of colonization of Vibrio cholerae strain C6709 and its isogenic knockout mutants in mouse intestine. A summary of the data, represented as mean Ϯ SD (n ϭ 10 mice per group), is shown. Variables were compared for significance using two-way analysis of variance and the Bonfer- roni test (*, P value between 0.01 and 0.05; **, P value between 0.01 to 0.001). FIG 5 Host cell death induced by OMVs from V. cholerae strains. (A) Flow cytometric analysis of cell death induced by V. cholerae OMVs. The graphical displays show data from a representative experiment. In each display the lower right quadrant represents apoptotic cells (annexin Vϩ /PIϪ ), and the upper left and right quadrants represent necrotic cells (annexin Vϩ /PIϩ and annexin VϪ /PIϩ fractions). Upper panel, untreated Int407 cells and Int407 cells treated with 20 ␮g/ml and 40 ␮g/ml of OMVs from V. cholerae strain C6709. Lower panel, Int407 cells treated with 20 ␮g/ml and 40 ␮g/ml OMVs from isogenic protease mutants of the C6709 and Comp VC1649 strains. (B) Results from three independent flow cytometry experiments. Data are represented as mean (ϮSD) percentage of dead cells after treatment with 20 and 40 ␮g/ml of OMVs from V. cholerae strains (n ϭ 3). (C) Fractions of apoptotic and necrotic cells of the same experiment. Data are represented as mean (ϮSD) percentages of apoptotic and necrotic cells (n ϭ 3). Mondal et al. 1486 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 10. purified earlier by Ghosh et al. in the presence of the metallopro- tease inhibitor EDTA, whereas in its absence the processed 35-kDa form is purified from culture supernatant (8). The protease un- dergoes several steps of processing, including cleavage of the signal peptide and further processing of the N terminus and C terminus to generate the 35-kDa protein (4). The presence of the 45-kDa form of HAP within the C6709 OMVs could be due to its lu- minal localization within the vesicles. Earlier studies have shown that luminal localization of proteins within OMVs pro- tects them from proteolytic digestions (40). Therefore, it may be suggested that the 45-kDa HAP is protected within the OMVs from further processing. OMVs have been shown to deliver different proteins and tox- ins into host cells in an active form (40). Several studies have shown that toxins such as CT, RTX, VCC, and metalloprotease PrtV in V. cholerae are transported to host cells through OMVs in a biologically active form (19, 20, 41). Our present study showed that both HAP- and VesC-associated OMVs are internalized into human intestinal epithelial cells (Int407) in a protease-indepen- dent manner. Confocal microscopic images revealed that these OMVs may be internalized into host cells through lipid rafts, as vesicular endocytosis was completely inhibited by M␤CD and nystatin. The proteases in OMVs are transported in biologically active forms. OMV-associated HAP induces dose-dependent morpho- logical changes in Int407 cells similar to those shown by purified HAP in our earlier study (8). HAP has been shown to proteolyti- cally activate El Tor hemolysin/cytolysin by nicking at its N-ter- minal peptide to generate the 65-kDa mature form (7). Our results showed that OMV-associated HAP also digests the 79-kDa pro- hemolysin to generate 65-kDa and 50-kDa mature forms. The 50-kDa mature hemolysin is a C-terminally processed, identically truncated monomer of the 65-kDa mature hemolysin reported by Ekigai et al. in V. cholerae O1 strains (42). The roles of OMV-associated proteases in pathogenesis were studied in the mouse ileal loop (MIL) model. This is the first study to show fluid accumulation in mouse ileal loops caused by V. cholerae OMV-associated proteases. HAP-associated OMVs showed an enterotoxic response in the MIL assay. VesC-associated CHA6.8 ⌬prtV OMVs showed a visible hemorrhagic fluid re- sponse in MILs. Histopathological analysis of the mice ileum showed grossly disrupted villi with hemorrhage in all layers of the mucosa, which was significantly reduced upon treatment with CHA6. 8⌬prtV ⌬VC1649 OMVs. These effects were restored with OMVs from the VC1649 complemented strain. A similar hemor- rhagic fluid response was observed with partially purified VesC and culture supernatants from CHA6.8 ⌬prtV in rabbit ileal loop (RIL) assay by Syngkon et al. They showed significant reduction of hemorrhagic fluid when inhibited with PMSF (10). This hemor- rhagic fluid accumulation was not due to cholera toxin, as CT- FIG 7 IL-8 induction by OMVs of V. cholerae O1 strains in the T84 cell line. (A) Comparative analysis of IL-8 secretion from T84 cells coincubated with OMVs from V. cholerae O1 El Tor strain C6709, its isogenic protease mutant strains CHA6.8, CHA6.8 ⌬prtV, and CHA6.8 ⌬prtV ⌬VC1649, and CHA6.8 ⌬prtV ⌬VC1649::VC1649 (Comp VC1649) determined by ELISA. Data represent mean Ϯ SD from three independent experiments performed under similar condi- tions. Variables were compared for significance using two-way analysis of variance and the Bonferroni test (*, P value between 0.01 and0.05; **, P value between 0.01 and 0.001). (B) IL-8 production from T84 cells was analyzed at the mRNA and protein levels by RT-PCR and resolved in a 2% agarose gel. IL-8 expression in T84 cells treated with OMVs from O1 El Tor strain C6709 and its protease knockout mutant strains CHA6.8, CHA6.8 ⌬prtV, CHA6.8 ⌬prtV ⌬VC1649, and Comp VC1649 is displayed in lanes 2 to 5, respectively, and the untreated control is in lane 1. Densitometric quantification in densitometric units (DU) for IL-8 after normalization to GAPDH is shown below the agarose gel. OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1487Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 11. bead ELISA showed the absence of CT in V. cholerae OMVs (Fig. 4C). The OMV-free culture supernatants also showed the absence of CT in bead ELISA (data not shown). This may be due to differ- ent conditions for production of CT and HAP. Benitez et al. showed that growth conditions that favor production of HAP downregulate CT and TCP and vice versa (43). OMV-associated proteases of V. cholerae induced cytotoxic ef- fects in Int407 cells in a dose-dependent manner. C6709 OMVs induced an apoptotic response in Int407 cells. Thay et al. have shown that membrane-derived vesicles from Staphylococcus au- reus induce apoptosis in a dose-dependent manner by an ␣-toxin- dependent mechanism (29). However, the apoptotic cell popula- tion was not significantly reduced when treated with CHA6.8 OMVs. CHA6.8 OMVs induced a greater cytotoxic response with an increase in necrotic cells compared to HAP-associated C6709 OMVs. Studies have shown that there is an increased virulence in hapA-deleted V. cholerae strains. Benitez et al. showed that inac- tivation of hapA results in increased intestinal colonization and adherence of V. cholerae in human intestinal epithelial cells (44). Zhou et al. also reported that hapA-defective cholera vaccine can- didate strains showed reactogenicity. Culture supernatants of these strains can stimulate IL-8 secretion from T-84 cells (45). Therefore, it may be suggested that the presence of metallopro- tease PrtV in CHA6.8 OMVs may be responsible for increased OMV-mediated cytotoxicity. However, earlier studies have shown that PrtV modulates IL-8 secretion from T84 cells (46). Interest- ingly, when cells were treated with CHA6.8 ⌬prtV, a shift from an apoptotic population to a necrotic population was observed. In CHA6.8 ⌬prtV ⌬VC1649 OMVs, the cytotoxicity was significantly reduced, and increased cytotoxicity in Int407 cells was restored with OMVs from the VC1649 complemented strain. These results are very similar to the effect observed with OMVs on MIL assay. Earlier studies on the pathogenicity of OMVs in bacterial in- fections have shown the role of lipopolysaccharide (LPS)- and OMV pathogen-associated molecular patterns (47, 48). LPS in the OMVs is sensed by the Toll-like receptor 4 (TLR4) complex to trigger inflammatory responses (49). OMVs isolated from Salmo- nella enterica serovar Typhimurium activate macrophages and dendritic cells to increase levels of surface major histocompatibil- ity complex (MHC) class II expression as well as the production of the proinflammatory mediators tumor necrosis factor alpha (TNF-␣) and interleukin-12 (IL-12) (50). Helicobacter pylori and Pseudomonas aeruginosa OMVs show a proinflammatory re- sponse by IL-8 secretion (51, 52). IL-8 is a major proinflammatory cytokine which act as a potent chemoattractant for polymorpho- nuclear leukocytes (PMN) and recruits them into infected sites of the epithelial layer, which results in opening of epithelial tight junctions (53). The IL-8 response of Vibrio cholerae OMVs was shown by Chatterjee and Chaudhuri. They showed that OMVs from classical V. cholerae strain O395 induce IL-8 secretion from intestinal epithelial cells, which activates dendritic cells to pro- mote T-cell polarization (54). However, our results showed that HAP-associated OMVs from El Tor strain C6709 failed to induce significant IL-8 secretion from T84 cells. Earlier studies have also shown that HAP does not induce IL-8 secretion from intestinal epithelial cells (45). OMVs from ⌬hapA strain CHA6.8 showed ϳ10-fold-increased IL-8 secretion compared to untreated con- trols, whereas IL-8 secretion was increased ϳ25-fold upon treat- ment with CHA6.8 ⌬prtV OMVs. The greater IL-8 response by OMVs from the ⌬hapA ⌬prtV double mutant strain could be due to the absence of PrtV, as it has been reported to reduce IL-8 secretion from intestinal epithelial cells (46). This increased IL-8 secretion by CHA6.8 ⌬prtV was significantly reduced upon treat- ment with OMVs from the VesC mutant strain. Interestingly, OMVs from Comp VC1649 showed significant induction of IL-8 secretion. These results show that VesC within the V. cholerae OMVs is responsible for the increased IL-8 response in T84 cells. Our results revealed 3-fold-increased colonization of the CHA6.8 ⌬prtV strain compared to the VesC-deleted CHA6.8⌬prtV⌬VC1649 strain in adult mice. Interestingly in- creased colonization of V. cholerae was restored with the VC1649 complemented strain. Earlier, Silva et al. (59) also reported an increased colonization of hapA mutant strains in infant mouse model. However, Sikora et al. reported that VesC is not responsi- ble for colonization of V. cholerae in infant mouse model (38). Our results showed that VesC may play an important role in coloniza- tion in adult mice. OMVs have recently been used to develop acellular vaccines for several diseases related to Gram-negative bacteria. The most suc- cessful use of OMVs as a vaccine candidate has been against sero- group B Neisseria meningitides infection, for which over 55 million doses have been administered to date against serogroup B infec- tions (55). Schild et al. showed that OMVs from V. cholerae can generate a protective immune response against cholera infection in the adult mouse model (25). However, our results suggest that the presence of proteases in the OMVs may affect the use of OMVs as a vaccine candidate for cholera. In conclusion, we showed that the proteases HAP and VesC are secreted in association with V. cholerae OMVs in an active form. OMV-associated VesC induces a hemorrhagic response in the MIL assay and a proinflammatory response in human cultured intestinal epithelial cells, and VesC may play an important role in colonization in the adult mouse model. ACKNOWLEDGMENTS This work was supported by intramural funding from the Indian Council of Medical Research. A.M. was supported by a fellowship from the Indian Council of Medical Research. We thank Somnath Chatterjee, confocal microscope operator, NICED central facility (Carl Zeiss, India), for technical help with confocal micro- scopic studies, Somdatta Chatterjee, Division of Bacteriology, NICED, for crucial assistance with VC1649 complementation, and Amarshi Mukher- jee, Division of Biochemistry, NICED, for providing purified hemolysin. REFERENCES 1. Farmer P, Almazor CP, Bahnsen ET, Barry D, Bazile J, Bloom BR, Bose N, Brewer T, Calderwood SB, Clemens JD, Cravioto A, Eustache E, Jérôme G, Gupta N, Harris JB, Hiatt HH, Holstein C, Hotez PJ, Ivers LC, Kerry VB, Koenig SP, Larocque RC, Léandre F, Lambert W, Lyon E, Mekalanos JJ, Mukherjee JS, Oswald C, Pape JW, Gretchko Prosper A, Rabinovich R, Raymonville M, Réjouit JR, Ronan LJ, Rosenberg ML, Ryan ET, Sachs JD, Sack DA, Surena C, Suri AA, Ternier R, Waldor MK, Walton D, Weigel JL. 2011. Meeting cholera’s challenge to Haiti and the world: a joint statement on cholera prevention and care. PLoS Negl Trop Dis 5:e1145. http://dx.doi.org/10.1371/journal.pntd.0001145. 2. Faruque SM, Albert MJ, Mekalanos JJ. 1998. Epidemiology, genetics, and ecology of toxigenic Vibrio cholerae. Microbiol Mol Biol Rev 62:1301– 1314. 3. Miyoshi S. 2013. Extracellular proteolytic enzymes produced by human pathogenic Vibrio species. Front Microbiol 4:339. http://dx.doi.org/10 .3389/fmicb.2013.00339. 4. Häse CC, Finkelstein RA. 1991. Cloning and nucleotide sequence of the Vibrio cholerae hemagglutinin/protease (HA/protease) gene and con- struction of an HA/protease-negative strain. J Bacteriol 173:3311–3317. Mondal et al. 1488 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 12. 5. Finkelstein RA, Boesman-Finkelstein M, Chang Y, Häse CC. 1992. Vibrio cholerae hemagglutinin/protease, colonial variation, virulence, and detachment. Infect Immun 60:472–478. 6. Booth BA, Boesman-Finkelstein M, Finkelstein RA. 1984. Vibrio chol- erae hemagglutinin/protease nicks cholera enterotoxin. Infect Immun 45: 558–560. 7. Nagamune K, Yamamoto K, Honda T. 1997. Intramolecular chaperone activity of the pro-region of Vibrio cholerae El Tor cytolysin. J Biol Chem 272:1338–1343. http://dx.doi.org/10.1074/jbc.272.2.1338. 8. Ghosh A, Saha DR, Hoque KM, Asakuna M, Yamasaki S, Koley H, Das SS, Chakrabarti MK, Pal A. 2006. Enterotoxigenicity of mature 45- kilodalton and processed 35-kilodalton forms of hemagglutinin protease purified from a cholera toxin gene-negative Vibrio cholerae non-O1, non- O139 strain. Infect Immun 74:2937–2946. http://dx.doi.org/10.1128/IAI .74.5.2937-2946.2006. 9. Vaitkevicius K, Lindmark B, Ou G, Song T, Toma C, Iwanaga M, Zhu J, Andersson A, Hammarström ML, Tuck S, Wai SN. 2006. A Vibrio cholerae protease needed for killing of Caenorhabditis elegans has a role in protection from natural predator grazing. Proc Natl Acad Sci U S A 103: 9280–9285. http://dx.doi.org/10.1073/pnas.0601754103. 10. Syngkon A, Elluri S, Koley H, Rompikunata PK, Saha DR, Chakrabarti MK, Bhadra RK, Wai SN, Pal A. 2010. Studies on a novel serine protease of a ⌬hapA⌬prtV Vibrio cholerae O1 strain and its role in haemorrhagic response in the rabbit ileal loop model. PLoS One 5:e13122. http://dx.doi .org/10.1371/journal.pone.0013122. 11. Pugsley AP. 1993. The complete general secretory pathway in gram- negative bacteria. Microbiol Rev 57:50–108. 12. Kuehn MJ, Kesty NC. 2005. Bacterial outer membrane vesicles and host pathogen interaction. Genes Dev 19:2645–2655. http://dx.doi.org/10 .1101/gad.1299905. 13. Wai SN, Lindmark B, Soderblom T, Takade A, Westermark M, Os- carsson J, Jass J, Richter-Dhalfors A, Mizunoe Y, Uhlin BE. 2003. Vesicle-mediated export and assembly of pore-forming oligomers of the enterobacterial ClyA cytotoxin. Cell 115:25–35. http://dx.doi.org/10.1016 /S0092-8674(03)00754-2. 14. Balsalobre C, Silvan JM, Berglund S, Mizunoe Y, Uhlin BE, Wai SN. 2006. Release of the type I secreted alpha-haemolysin via outer membrane vesicles from Escherichia coli. Mol Microbiol 59:99–112. http://dx.doi.org /10.1111/j.1365-2958.2005.04938.x. 15. Davis JM, Carvalho HM, Rasmussen SB, O’Brien AD. 2006. Cytotoxic necrotizing factor type 1 delivered by outer membrane vesicles of uro- pathogenic E. coli attenuates polymorphonuclear leukocyte antimicrobial activity and chemotaxis. Infect Immun 74:4401–4408. http://dx.doi.org /10.1128/IAI.00637-06. 16. Lindmark B, Rompikuntal PK, Vaitkevicius K, Song T, Mizunoe Y, Uhlin BE, Guerry P, Wai SN. 2009. Outer membrane vesicle-mediated release of cytolethal distending toxin (CDT) from Campylobacter jejuni. BMC Microbiol 9:220. http://dx.doi.org/10.1186/1471-2180-9-220. 17. Parker H, Chitcholtan K, Hampton MB, Keenan JI. 2010. Uptake of Helicobacter pylori outer membrane vesicles by gastric epithelial cells. In- fect Immun 78:5054–5061. http://dx.doi.org/10.1128/IAI.00299-10. 18. Bomberger JM, MacEachran DP, Coutermarsh BA, Ye S, O’Toole GA, Stanton BA. 2009. Long-distance delivery of bacterial virulence factors by Pseudomonas aeruginosa outer membrane vesicles. PLoS Pathog 5:e1000382. http://dx.doi.org/10.1371/journal.ppat.1000382. 19. Chatterjee D, Chaudhuri K. 2011. Association of cholera toxin with Vibrio cholerae outer membrane vesicles which are internalized by human intestinal epithelial cells. FEBS Lett 585:1357–1362. http://dx.doi.org/10 .1016/j.febslet.2011.04.017. 20. Elluri S, Enow C, Vdovikova S, Rompikunal P, Dongre M, Carlsson S, Pal A, Uhlin BE, Wai SN. 2014. Outer membrane vesicles mediate transport of biologically active Vibrio cholerae cytolysin (VCC) from V. cholerae strains. PLoS One 9:e106731. http://dx.doi.org/10.1371/journal .pone.0106731. 21. Rompikuntal PK, Vdovikova S, Duperthuy M, Jhonson TL, Ahlund M, Lundmark R, Oscarson J, Sandkvist M, Uhlin BE, Wai SN. 2015. Outer membrane vesicle mediated export of processed PrtV protease from Vibrio cholerae. PLoS One 10:e0134098. http://dx.doi.org/10.1371/journal .pone.0134098. 22. Altindis E, Fu Y, Mekalanos JJ. 2014. Proteomic analysis of Vibrio chol- erae outer membrane vesicles. Proc Natl Acad Sci U S A 111:E1548– E1556. http://dx.doi.org/10.1073/pnas.1403683111. 23. Gosink KK, Kobayashi R, Kawagishi I, Hase CC. 2002. Analyses of the roles of three cheA homologs in chemotaxis of Vibrio cholerae. J Bacteriol 184:1767–1771. http://dx.doi.org/10.1128/JB.184.6.1767-1771.2002. 24. Chen DJ, Osterrieder N, Metzger SM, Buckles E, Doody AM, DeLisa MP, Putnam D. 2010. Delivery of foreign antigens by engineered outer membrane vesicle vaccines. Proc Natl Acad Sci U S A 107:3099–3104. http://dx.doi.org/10.1073/pnas.0805532107. 25. Schild S, Nelson EJ, Camilli A. 2008. Immunization with Vibrio cholerae outer membrane vesicles induces protective immunity in mice. Infect Im- mun 76:4554–4563. http://dx.doi.org/10.1128/IAI.00532-08. 26. Cheng LW, Schneewind O. 2000. Yersinia enterocolitica TyeA, an intra- cellular regulator of the type III machinery, is required for specific target- ing of YopE, YopH, YopM, and YopN into the cytosol of eukaryotic cells. J Bacteriol 182:3183–3190. http://dx.doi.org/10.1128/JB.182.11.3183 -3190.2000. 27. Parker H, Chitcholtan K, Hampton MB, Keenan JI. 2010. Uptake of Helicobacter pylori outer membrane vesicles by gastric epithelial cells. In- fect Immun 72:5054–5061. http://dx.doi.org/10.1128/IAI.00299-10. 28. Sawasvirojwong S, Srimanote P, Chatsudthipong V, Muanprasat C. 2013. An adult mouse model of Vibrio cholerae induced diarrhea for study- ing pathogenesis and potential therapy of cholera. PLoS Negl Trop Dis 7:e2293. http://dx.doi.org/10.1371/journal.pntd.0002293. 29. Thay B, Wai SN, Oscarsson J. 2013. Staphylococcus aureus ␣-toxin de- pendent induction of host cell death by membrane derived vesicles. PLoS One 8:e54661. http://dx.doi.org/10.1371/journal.pone.0054661. 30. Oku Y, Uesaka Y, Hirayama T, Takeda Y. 1988. Development of highly sensitive bead-ELISA to detect bacterial protein toxins. Microbiol Immu- nol 32:807–816. http://dx.doi.org/10.1111/j.1348-0421.1988.tb01442.x. 31. Olivier V, Queen J, Satchell KJF. 2009. Successful small intestine colo- nization of adult mice by Vibrio cholerae requires ketamine anesthesia and accessory toxins. PLoS One 4:e7352. http://dx.doi.org/10.1371/journal .pone.0007352. 32. Chatterjee SN, Das J. 1967. Electron microscopic observations on the excretion of cell-wall material by Vibrio cholerae. J Gen Microbiol 49:1–11. http://dx.doi.org/10.1099/00221287-49-1-1. 33. Rodal SK, Skretting G, Garred F, Vilhardt B, Van Duers B, Sandvig. 1999. Extraction of cholesterol with methyl-beta-cyclodextrin perturbs formation of clathrin-coated endocytic vesicles. Mol Biol Cell 10:961–974. http://dx.doi.org/10.1091/mbc.10.4.961. 34. Bonnington KE, Kuehn MJ. 2014. Protein secretion and export via outer membrane vesicles. Biochim Biophys Acta 1843:1612–1619. http://dx.doi .org/10.1016/j.bbamcr.2013.12.011. 35. Sheikh IA, Koley H, Chakrabarti MK, Hoque KM. 2013. The Epac signalling pathway regulates ClϪ secretion via modulation of apical KCNN4c channels in diarrhea. J Biol Chem 288:20404–20415. http://dx .doi.org/10.1074/jbc.M113.467860. 36. Scott ME, Dossani ZY, Sandkvist M. 2001. Directed polar secretion of protease from single cells of Vibrio cholerae via the type II secretion path- way. Proc Natl Acad Sci U S A 98:13978–13983. http://dx.doi.org/10.1073 /pnas.241411198. 37. Young DB, Broadbent DA. 1982. Biochemical characterization of extra- cellular proteases from Vibrio cholerae. Infect Immun 37:875–883. 38. Sikora AE, Zielke RA, Lawrence DA, Andrews PC, Sandkvist M. 2011. Proteomic analysis of the Vibrio cholerae type II secretome reveals new proteins, including three related serine proteases. J Biol Chem 286:16555– 16566. http://dx.doi.org/10.1074/jbc.M110.211078. 39. Silva AJ, Pham K, Benitez JA. 2003. Hemaglutinnin/protease expression and mucin gel penetration in El Tor biotype Vibrio cholerae. Microbiology 149:1883–1891. http://dx.doi.org/10.1099/mic.0.26086-0. 40. Kulp A, Kuehn MJ. 2010. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev Microbiol 64:163–184. http: //dx.doi.org/10.1146/annurev.micro.091208.073413. 41. Boardman BK, Meehan BM, Fullner-Satchell KJ. 2007. Growth phase regulation of Vibrio cholerae RTX toxin export. J Bacteriol 189:1827–1835. http://dx.doi.org/10.1128/JB.01766-06. 42. Ikigai H, Ono T, Nakae T, Otsuru H, Shimamura T. 1999. Two form of Vibrio cholerae O1 El Tor hemolysin derived from identical precursor protein. Biochim Biophys Acta 1415:297–305. http://dx.doi.org/10.1016 /S0005-2736(98)00183-7. 43. Benitez JA, Silva AJ, Finkelstein RA. 2001. Environmental signals con- trolling production of hemagglutinin/protease in Vibrio cholerae. Infect Immun 69:6549–6553. http://dx.doi.org/10.1128/IAI.69.10.6549-6553 .2001. 44. Bénitez JA, Spelbrink RG, Silva A, Phillips TE, Stanley CM, Boesman- OMV-Associated Active Proteases in V. cholerae May 2016 Volume 84 Number 5 iai.asm.org 1489Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom
  • 13. Finkelstein M, Finkelstein RA. 1997. Adherence of Vibrio cholerae to cultured differentiated human intestinal cells: an in vitro colonization model. Infect Immun 65:3474–3477. 45. Zhou X, Gao DQ, Michalski J, Benitez JA, Kaper JB. 2004. Induction of interleukin-8 in T84 cells by Vibrio cholerae. Infect Immun 72:389–397. http://dx.doi.org/10.1128/IAI.72.1.389-397.2004. 46. Ou G, Rompikuntal PK, Bitar A, Lindmark B, Vaitkevicius K, Wai SN, Hammarström ML. 2009. Vibrio cholerae cytolysin causes an inflamma- tory response in human intestinal epithelial cells that is modulated by the PrtV protease. PLoS One 4:e7806. http://dx.doi.org/10.1371/journal.pone .0007806. 47. Whitmire WM, Garon CF. 1993. Specific and nonspecific responses of murine B cells to membrane blebs of Borrelia burgdoferi. Infect Immun 61:1460–1467. 48. Opal SM. 2007. The host response to endotoxin, antilipopolysaccaride strategies, and the management of severe sepsis. Int J Med Microbiol 297: 365–377. http://dx.doi.org/10.1016/j.ijmm.2007.03.006. 49. Ellis TN, Kuehn MJ. 2010. Virulence and immunomodulatory roles of bacterial outer membrane vesicles. Microbiol Mol Biol Rev 74:81–94. http: //dx.doi.org/10.1128/MMBR.00031-09. 50. Alaniz RC, Deatherage BL, Lara JC, Cookson BT. 2007. Membrane vesicles are immunogenic facsimiles of Salmonella typhimurium that po- tently activate dendritic cells, prime B and T cell responses, and stimulate protective immunity in vivo. J Immunol 179:7692–7701. http://dx.doi.org /10.4049/jimmunol.179.11.7692. 51. Bauman SJ, Kuehn MJ. 2006. Purification of outer membrane vesicles from Pseudomonas aeruginosa and their activation of an IL-8 response. Microbes Infect 8:2400–2408. http://dx.doi.org/10.1016/j.micinf.2006.05 .001. 52. Ismail S, Hampton MB, Keenan JI. 2003. Helicobacter pylori outer mem- brane vesicles modulate proliferation and interleukin-8 production by gastric epithelial cells. Infect Immun 71:5670–5675. http://dx.doi.org/10 .1128/IAI.71.10.5670-5675.2003. 53. McCormick BA, Siber AM, Maurelli AT. 1998. Requirement of the Shigella flexneri virulence plasmid in the ability to induce trafficking of neutrophils across polarized monolayers of the intestinal epithelium. In- fect Immun 66:4237–4243. 54. Chatterjee D, Chaudhuri K. 2013. Vibrio cholera O395 outer membrane vesicles modulate intestinal epithelial cells in a NOD1 dependent manner and induces dendritic cell-mediated Th2/Th17 responses. J Biol Chem 288:4299–4309. http://dx.doi.org/10.1074/jbc.M112.408302. 55. Holst J, Martin D, Arnold R, Huergo CC, Oster P, O’Hallahhan J, Rosenqvist E. 2009. Properties and clinical performance of vaccines con- taining outer membrane vesicles from Neisseria meningitides. Vaccine 27: B3–B12. http://dx.doi.org/10.1016/j.vaccine.2009.04.071. 56. Haralalka S, Nandi S, Bhadra RK. 2003. Mutation in the relA gene of Vibrio cholerae affects in vitro and in vivo expression of virulence factors. J Bacteriol 185:4672–4682. http://dx.doi.org/10.1128/JB.185.16.4672 -4682.2003. 57. Jung HC, Eckmann L, Yang SK, Panja A, Fierer J, Morzycka- Wroblewska E, Kagnoff MF. 1995. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bac- terial invasion. J Clin Invest 95:55–65. http://dx.doi.org/10.1172 /JCI117676. 58. Sarkar M, Chaudhuri K. 2004. Association of adherence and motility in interleukin 8 induction in human intestinal epithelial cells by Vibrio chol- erae. Microbes Infect 6:676–685. http://dx.doi.org/10.1016/j.micinf.2004 .02.018. 59. Silva AJ, Leitch GJ, Camilli A, Benitez JA. 2006. Contribution of hem- agglutinin/protease and motility to the pathogenesis of El Tor biotype cholera. Infect Immun 74:2072–2079. http://dx.doi.org/10.1128/IAI.74.4 .2072-2079.2006. Mondal et al. 1490 iai.asm.org May 2016 Volume 84 Number 5Infection and Immunity onApril22,2016byUniversityofCalifornia,Berkeleyhttp://iai.asm.org/Downloadedfrom